1
|
Wu XQ, Su N, Fei Z, Fei F. Homer signaling pathways as effective therapeutic targets for ischemic and traumatic brain injuries and retinal lesions. Neural Regen Res 2021; 17:1454-1461. [PMID: 34916418 PMCID: PMC8771115 DOI: 10.4103/1673-5374.330588] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Ischemic and traumatic insults to the central nervous system account for most serious acute and fatal brain injuries and are usually characterized by primary and secondary damage. Secondary damage presents the greatest challenge for medical staff; however, there are currently few effective therapeutic targets for secondary damage. Homer proteins are postsynaptic scaffolding proteins that have been implicated in ischemic and traumatic insults to the central nervous system. Homer signaling can exert either positive or negative effects during such insults, depending on the specific subtype of Homer protein. Homer 1b/c couples with other proteins to form postsynaptic densities, which form the basis of synaptic transmission, while Homer1a expression can be induced by harmful external factors. Homer 1c is used as a unique biomarker to reveal alterations in synaptic connectivity before and during the early stages of apoptosis in retinal ganglion cells, mediated or affected by extracellular or intracellular signaling or cytoskeletal processes. This review summarizes the structural features, related signaling pathways, and diverse roles of Homer proteins in physiological and pathological processes. Upregulating Homer1a or downregulating Homer1b/c may play a neuroprotective role in secondary brain injuries. Homer also plays an important role in the formation of photoreceptor synapses. These findings confirm the neuroprotective effects of Homer, and support the future design of therapeutic drug targets or gene therapies for ischemic and traumatic brain injuries and retinal disorders based on Homer proteins.
Collapse
Affiliation(s)
- Xiu-Quan Wu
- Department of Neurosurgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Ning Su
- Department of Radiation Oncology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Fei Fei
- Department of Ophthalmology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
2
|
Clifton NE, Trent S, Thomas KL, Hall J. Regulation and Function of Activity-Dependent Homer in Synaptic Plasticity. MOLECULAR NEUROPSYCHIATRY 2019; 5:147-161. [PMID: 31312636 DOI: 10.1159/000500267] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/09/2019] [Indexed: 12/22/2022]
Abstract
Alterations in synaptic signaling and plasticity occur during the refinement of neural circuits over the course of development and the adult processes of learning and memory. Synaptic plasticity requires the rearrangement of protein complexes in the postsynaptic density (PSD), trafficking of receptors and ion channels and the synthesis of new proteins. Activity-induced short Homer proteins, Homer1a and Ania-3, are recruited to active excitatory synapses, where they act as dominant negative regulators of constitutively expressed, longer Homer isoforms. The expression of Homer1a and Ania-3 initiates critical processes of PSD remodeling, the modulation of glutamate receptor-mediated functions, and the regulation of calcium signaling. Together, available data support the view that Homer1a and Ania-3 are responsible for the selective, transient destabilization of postsynaptic signaling complexes to facilitate plasticity of the excitatory synapse. The interruption of activity-dependent Homer proteins disrupts disease-relevant processes and leads to memory impairments, reflecting their likely contribution to neurological disorders.
Collapse
Affiliation(s)
- Nicholas E Clifton
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom.,MRC Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Simon Trent
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Kerrie L Thomas
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom.,School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Jeremy Hall
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom.,MRC Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
3
|
El-Sitt S, Soueid J, Al Ali J, Makoukji J, Makhoul NJ, Harati H, Boustany RM. Developmental Comparison of Ceramide in Wild-Type and Cln3 Δex7/8 Mouse Brains and Sera. Front Neurol 2019; 10:128. [PMID: 30837943 PMCID: PMC6389635 DOI: 10.3389/fneur.2019.00128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/31/2019] [Indexed: 11/24/2022] Open
Abstract
CLN3 disease is a neurodevelopmental disease leading to early visual failure, motor decline, and death. CLN3 pathogenesis has been linked to dysregulation of ceramide, a key intracellular messenger impacting various biological functions. Ceramide is upregulated in brains of CLN3 patients and activates apoptosis. Ceramide levels over the lifespan of WT and Cln3Δex7/8 mice were measured using the DGK assay. Ceramide subspecies were determined by LC-MS. Ceramide synthesis enzymes and pre- and post-synaptic mRNA expression was measured in Cln3Δex7/8 and normal mouse brains. Neuronal cell death was established by PARP cleavage and Caspases 3/6/9 and cytochrome C mRNA expression in Cln3Δex7/8 and normal mouse brains. In WT mouse, a ceramide peak was noted at 3 weeks of age. The absence of this peak in Cln3Δex7/8 mice might be related to early disease pathogenesis. Increase of ceramide in Cln3Δex7/8 mouse brain at 24 weeks of age precedes neuronal apoptosis. The correlation between serum and brain ceramide in WT mice, and dysregulation of ceramide in serum and brain of Cln3Δex7/8 mice, and the significant increase in ceramide in Cln3Δex7/8 mouse brains and sera argue for use of easily accessible serum ceramide levels to track response to novel therapies in human CLN3 disease.
Collapse
Affiliation(s)
- Sally El-Sitt
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Jihane Soueid
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Jamal Al Ali
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Joelle Makoukji
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nadine J Makhoul
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hayat Harati
- Neuroscience Research Centre, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Rose-Mary Boustany
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
4
|
Shao X, Hu Q, Chen S, Wang Q, Xu P, Jiang X. Ghrelin Ameliorates Traumatic Brain Injury by Down-Regulating bFGF and FGF-BP. Front Neurosci 2018; 12:445. [PMID: 30026681 PMCID: PMC6041414 DOI: 10.3389/fnins.2018.00445] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/12/2018] [Indexed: 12/15/2022] Open
Abstract
Traumatic brain injury (TBI) is a primary cause of disability and mortality. Ghrelin, a gastrointestinal hormone, has been found to have protective effects for the brain, but the molecular mechanism of these neuroprotective effects of ghrelin remains unclear. In this study, an electronic cortical contusion impactor was used to establish a rat TBI model and we investigated the effect of ghrelin on brain repair by neurological severity score and histological examination. An antibody array was employed to uncover the molecular mechanism of ghrelin’s neuroprotective effects by determining the alterations of multiple proteins in the brain cortex. As a result, ghrelin attenuated brain injury and promoted brain functional recovery. After TBI, 13 proteins were up-regulated in the brain cortex, while basic fibroblast growth factor (bFGF) and fibroblast growth factor-binding protein (FGF-BP) were down-regulated after ghrelin treatment. It is known that bFGF can induce angiogenesis in the brain and accelerate wound healing, which can be further enhanced by FGF-BP. Based on the previous studies, it is hypothesized that the exogenous ghrelin curing TBI might cause the closure of bFGF and FGF-BP functions on wound healing, or ghrelin might exert the neuroprotective effects by competitively inhibiting bFGF/FGF-BP-induced neovascularization. Whether the combinational administration of ghrelin and bFGF/FGF-BP can enhance or weaken the therapeutic effect on TBI requires further research.
Collapse
Affiliation(s)
- Xuefei Shao
- Department of Neurosurgery, Yi-Ji Shan Hospital of Wannan Medical College, Wuhu, China
| | - Qianxin Hu
- Department of Neurosurgery, Yi-Ji Shan Hospital of Wannan Medical College, Wuhu, China
| | - Sansong Chen
- Department of Neurosurgery, Yi-Ji Shan Hospital of Wannan Medical College, Wuhu, China
| | - Qifu Wang
- Department of Neurosurgery, Yi-Ji Shan Hospital of Wannan Medical College, Wuhu, China
| | - Pengcheng Xu
- Department of Neurosurgery, Yi-Ji Shan Hospital of Wannan Medical College, Wuhu, China
| | - Xiaochun Jiang
- Department of Neurosurgery, Yi-Ji Shan Hospital of Wannan Medical College, Wuhu, China
| |
Collapse
|
5
|
Tomasetti C, Iasevoli F, Buonaguro EF, De Berardis D, Fornaro M, Fiengo ALC, Martinotti G, Orsolini L, Valchera A, Di Giannantonio M, de Bartolomeis A. Treating the Synapse in Major Psychiatric Disorders: The Role of Postsynaptic Density Network in Dopamine-Glutamate Interplay and Psychopharmacologic Drugs Molecular Actions. Int J Mol Sci 2017; 18:E135. [PMID: 28085108 PMCID: PMC5297768 DOI: 10.3390/ijms18010135] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 12/25/2016] [Accepted: 01/08/2017] [Indexed: 12/20/2022] Open
Abstract
Dopamine-glutamate interplay dysfunctions have been suggested as pathophysiological key determinants of major psychotic disorders, above all schizophrenia and mood disorders. For the most part, synaptic interactions between dopamine and glutamate signaling pathways take part in the postsynaptic density, a specialized ultrastructure localized under the membrane of glutamatergic excitatory synapses. Multiple proteins, with the role of adaptors, regulators, effectors, and scaffolds compose the postsynaptic density network. They form structural and functional crossroads where multiple signals, starting at membrane receptors, are received, elaborated, integrated, and routed to appropriate nuclear targets. Moreover, transductional pathways belonging to different receptors may be functionally interconnected through postsynaptic density molecules. Several studies have demonstrated that psychopharmacologic drugs may differentially affect the expression and function of postsynaptic genes and proteins, depending upon the peculiar receptor profile of each compound. Thus, through postsynaptic network modulation, these drugs may induce dopamine-glutamate synaptic remodeling, which is at the basis of their long-term physiologic effects. In this review, we will discuss the role of postsynaptic proteins in dopamine-glutamate signals integration, as well as the peculiar impact of different psychotropic drugs used in clinical practice on postsynaptic remodeling, thereby trying to point out the possible future molecular targets of "synapse-based" psychiatric therapeutic strategies.
Collapse
Affiliation(s)
- Carmine Tomasetti
- NHS, Department of Mental Health ASL Teramo, Psychiatric Service of Diagnosis and Treatment, Hospital "Maria SS dello Splendore", 641021 Giulianova, Italy.
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Reproductive and Odontostomatogical Sciences, University of Naples "Federico II", 80131 Napoli, Italy.
- Polyedra Research Group, 64100 Teramo, Italy.
| | - Felice Iasevoli
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Reproductive and Odontostomatogical Sciences, University of Naples "Federico II", 80131 Napoli, Italy.
- Polyedra Research Group, 64100 Teramo, Italy.
| | - Elisabetta Filomena Buonaguro
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Reproductive and Odontostomatogical Sciences, University of Naples "Federico II", 80131 Napoli, Italy.
- Polyedra Research Group, 64100 Teramo, Italy.
| | - Domenico De Berardis
- Polyedra Research Group, 64100 Teramo, Italy.
- NHS, Department of Mental Health ASL Teramo, Psychiatric Service of Diagnosis and Treatment, Hospital "G. Mazzini", 64100 Teramo, Italy.
- Department of Neuroscience and Imaging, University "G. d'Annunzio", 66100 Chieti, Italy.
| | - Michele Fornaro
- Polyedra Research Group, 64100 Teramo, Italy.
- New York State Psychiatric Institute, Columbia University, New York, NY 10027, USA.
| | | | - Giovanni Martinotti
- Polyedra Research Group, 64100 Teramo, Italy.
- Department of Neuroscience and Imaging, University "G. d'Annunzio", 66100 Chieti, Italy.
| | - Laura Orsolini
- Polyedra Research Group, 64100 Teramo, Italy.
- Casa di Cura Villa San Giuseppe, 63100 Ascoli Piceno, Italy.
| | - Alessandro Valchera
- Polyedra Research Group, 64100 Teramo, Italy.
- Casa di Cura Villa San Giuseppe, 63100 Ascoli Piceno, Italy.
| | | | - Andrea de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Reproductive and Odontostomatogical Sciences, University of Naples "Federico II", 80131 Napoli, Italy.
| |
Collapse
|
6
|
Wen X, Chen X, Chen S, Tan Y, Rong F, Zhu J, Ma W. Influence of SKF38393 on changes of gene profile in rat prefrontal cortex during chronic paradoxical sleep deprivation. Behav Brain Res 2016; 304:60-6. [PMID: 26851556 DOI: 10.1016/j.bbr.2016.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 01/27/2016] [Accepted: 02/01/2016] [Indexed: 11/17/2022]
Abstract
Chronic paradoxical sleep deprivation (CSD) can induce dramatic physiological and neurofunctional changes in rats, including decreased body weight, reduced learning and memory, and declined locomotor function. SKF38393, a dopamine D1 receptor agonist, can reverse the above damages. However, the mechanism of CSD syndrome and reversal role of SKF38393 remains largely unexplained. To preliminarily elucidate the mechanism of the neural dysfunction caused by CSD, in the present study we use gene chips to examine the expression profile of more than 28,000 transcripts in the prefrontal cortex (PFC). Rats were sleep deprived by modified multi-platform method for 3 weeks. Totally 59 transcripts showed differential expressions in CSD group in contrast to controls; they included transcripts coding for caffeine metabolism, circadian rhythm, drug metabolism and some amino acid metabolism pathway. Among the 59 transcripts, 39 increased their expression and 20 decreased. Two transcripts can be specifically reversed with SKF38393, one of them is Homer1, which is related to 20 functional classifications and coding for Glutamatergic synapse pathway. Our findings in the present study indicate that long-term sleep deprivation may trigger the changes of some certain functions and pathways in the PFC, and lead to the dysfunction of this advanced neuron, and the activation of D1 receptor by SKF38393 might ameliorate these changes via modulation of some transcripts such as Homer1, which is involved in the Ca(2+) pathway and MAPK pathway related to Glutamatergic synapse pathway.
Collapse
Affiliation(s)
- Xiaosa Wen
- Department of Enviromental Hygiene, Faculty of Tropical Medicine and Public Health, Second Military Medical University, Shanghai 200433, China; Minhang District Center for Disease Control and Prevention, Shanghai 201101, China
| | - Xinmin Chen
- Department of Enviromental Hygiene, Faculty of Tropical Medicine and Public Health, Second Military Medical University, Shanghai 200433, China
| | - Si Chen
- Department of Enviromental Hygiene, Faculty of Tropical Medicine and Public Health, Second Military Medical University, Shanghai 200433, China
| | - Yue Tan
- Surgical Department, Tangshan Gongren Hospital, Tangshan 063000, China
| | - Fei Rong
- Department of Enviromental Hygiene, Faculty of Tropical Medicine and Public Health, Second Military Medical University, Shanghai 200433, China
| | - Jiangbo Zhu
- Department of Enviromental Hygiene, Faculty of Tropical Medicine and Public Health, Second Military Medical University, Shanghai 200433, China.
| | - Wenling Ma
- Department of Enviromental Hygiene, Faculty of Tropical Medicine and Public Health, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
7
|
Hou PF, Liu ZH, Li N, Cheng WJ, Guo SW. Knockdown of STIM1 improves neuronal survival after traumatic neuronal injury through regulating mGluR1-dependent Ca(2+) signaling in mouse cortical neurons. Cell Mol Neurobiol 2015; 35:283-92. [PMID: 25304289 PMCID: PMC11486307 DOI: 10.1007/s10571-014-0123-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 10/05/2014] [Indexed: 01/05/2023]
Abstract
Activation of glutamate receptors and followed increase of intracellular calcium concentration is a key pathological mechanism involved in secondary neuronal injury after traumatic brain injury (TBI). Stromal interaction molecule (STIM) proteins are considered to be important players in regulating neuronal Ca(2+) homeostasis under normal aging and pathological conditions. Here, we investigated the role of STIM1 in regulating metabotropic glutamate receptor 1 (mGluR1)-related Ca(2+) signaling and neuronal survival by using an in vitro traumatic neuronal injury (TNI) model. The expression of STIM1 was significantly increased at both mRNA and protein levels after TNI. Down-regulation of STIM1 by specific small interfere RNA significantly preserved neuronal viability, decreased lactate dehydrogenase release, and inhibited apoptotic cell death after traumatic injury. Moreover, knockdown of STIM1 significantly alleviated the mGluR1-related increase of cytoplasmic Ca(2+) levels after TNI. By analyzing Ca(2+) imaging in Ca(2+)-free conditions, we demonstrated that the mGluR1-dependent inositol trisphosphate receptor and/or ryanodine receptor-mediated Ca(2+) release from the endoplasmic reticulum after TNI is strongly attenuated in the absence of STIM1. Together, our results demonstrate that in the mammalian nervous system, STIM1 is a key regulator of mGluR1-dependent Ca(2+) signaling and knockdown of STIM1 might be an effective intervention target in TBI.
Collapse
Affiliation(s)
- Peng-Fei Hou
- Department of Neurosurgery, The First Affiliated Hospital of Xi’an Jiaotong University, 277 West Yanta Road, Xi’an, 710061 Shaanxi China
- Department of Neurosurgery, Ninth Hospital of Xi’an, Xi’an, 710054 Shaanxi China
| | - Zhan-Hui Liu
- Department of Neurosurgery, Ninth Hospital of Xi’an, Xi’an, 710054 Shaanxi China
| | - Nan Li
- Department of Neurosurgery, The First Affiliated Hospital of Xi’an Jiaotong University, 277 West Yanta Road, Xi’an, 710061 Shaanxi China
- Department of Neurosurgery, Xi’an Chidren’s Hospital, Xi’an, 710043 Shaanxi China
| | - Wen-Jia Cheng
- Department of Pathology, Ninth Hospital of Xi’an, Xi’an, 710054 Shaanxi China
| | - Shi-Wen Guo
- Department of Neurosurgery, The First Affiliated Hospital of Xi’an Jiaotong University, 277 West Yanta Road, Xi’an, 710061 Shaanxi China
| |
Collapse
|
8
|
Huang W, Liu X, Fei Z, Zhang Y, Yang J. Down-regulation of Homer1b/c expression protects cultured neurons after traumatic injury. Neural Regen Res 2014; 7:2176-81. [PMID: 25538737 PMCID: PMC4268715 DOI: 10.3969/j.issn.1673-5374.2012.028.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 09/24/2012] [Indexed: 11/18/2022] Open
Abstract
Activation of metabotropic glutamate receptor 1a aggravates traumatic brain injury. The constitutively expressed protein Homer1b/c participates in delivering and anchoring metabotropic glutamate receptors in neurons. Here, we aimed to verify whether down-regulation of Homer1b/c by RNA interference could protect cultured rat cortical neurons from traumatic injury. We showed that 36 hours after transfection of Homer1b/c small interfering RNA, metabotropic glutamate receptor 1a was present only in the neuronal cytoplasm, but not in the dendrites. Calcium fluorescence intensity was also decreased significantly. Moreover, lactate dehydrogenase concentration was significantly decreased in Homer1b/c small interfering RNA-transfected cells compared with that in untransfected and control small interfering RNA-transfected cells 24 hours after traumatic neuronal injury. Our findings indicate that down-regulation of Homer1b/c could reduce metabotropic glutamate receptor 1a transfer from the cell body to the dendrite, relieve calcium overload, and protect neurons from traumatic injury.
Collapse
Affiliation(s)
- Weidong Huang
- Department of Neurosurgery, Shaanxi Provincial People's Hospital, Xi'an 710068, Shaanxi Province, China
| | - Xiaobin Liu
- Department of Neurosurgery, Shaanxi Provincial People's Hospital, Xi'an 710068, Shaanxi Province, China
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, the Fourth Military Medical University of Chinese PLA, Xi'an 710032, Shaanxi Province, China
| | - Yuelin Zhang
- Department of Neurosurgery, Shaanxi Provincial People's Hospital, Xi'an 710068, Shaanxi Province, China
| | - Jun Yang
- Department of Neurosurgery, Shaanxi Provincial People's Hospital, Xi'an 710068, Shaanxi Province, China
| |
Collapse
|
9
|
Lv MM, Cheng YC, Xiao ZB, Sun MY, Ren PC, Sun XD. Down-regulation of Homer1b/c attenuates group I metabotropic glutamate receptors dependent Ca²⁺ signaling through regulating endoplasmic reticulum Ca²⁺ release in PC12 cells. Biochem Biophys Res Commun 2014; 450:1568-74. [PMID: 25026550 DOI: 10.1016/j.bbrc.2014.07.044] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 07/07/2014] [Indexed: 10/25/2022]
Abstract
The molecular basis for group I metabotropic glutamate receptors (mGluR1 and 5) coupling to membrane ion channels and intracellular calcium pools is not fully understood. Homer is a family of post synaptic density proteins functionally and physically attached to target proteins at proline-rich sequences. In the present study, we demonstrate that Homer1b/c is constitutively expressed in PC12 cells, whereas Homer1a, the immediate early gene product, can be up-regulated by brain derived neurotrophic factor (BDNF) and glutamate. Knockdown of Homer1b/c using specific target small interfering RNA (siRNA) did not interfere the expression of mGluR1, mGluR5 and their downstream effectors, including inositol-1,4,5-trisphosphate receptors (IP3R), phospholipase C (PLC) and Gq proteins. By analyzing Ca(2+) imaging in PC12 cells, we demonstrated that Homer1b/c is an essential regulator of the Ca(2+) release from the endoplasmic reticulum (ER) induced by the activation of group I mGluRs, IP3R and ryanodine receptors (RyR). Furthermore, the group I mGluRs activation-dependent refilling of the Ca(2+) stores in both resting and depolarizing conditions were strongly attenuated in the absence of Homer1b/c. Together, our results demonstrate that in PC12 cells Homer1b/c is a regulator of group I mGluRs related Ca(2+) homeostasis that is essential for the maintenance of normal Ca(2+) levels in the ER.
Collapse
Affiliation(s)
- Miao-Miao Lv
- Department of Anesthesiology, Tangdu Hospital of the Fourth Military Medical University, Xi'an, Shaanxi 710032, China; Department of Anesthesiology, The 323 Hospital of PLA, Xi'an, Shaanxi 710054, China
| | - Yong-Chun Cheng
- Department of Anesthesiology, The Third Hospital of PLA, Baoji, Shaanxi 721004, China
| | - Zhi-Bin Xiao
- Department of Anesthesiology, The 323 Hospital of PLA, Xi'an, Shaanxi 710054, China
| | - Mei-Yan Sun
- Department of Anesthesiology, Tangdu Hospital of the Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Peng-Cheng Ren
- Department of Anesthesiology, Tangdu Hospital of the Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xu-De Sun
- Department of Anesthesiology, Tangdu Hospital of the Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
10
|
Fei F, Rao W, Zhang L, Chen BG, Li J, Fei Z, Chen Z. Downregulation of Homer1b/c improves neuronal survival after traumatic neuronal injury. Neuroscience 2014; 267:187-94. [PMID: 24607348 DOI: 10.1016/j.neuroscience.2014.02.037] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 02/04/2014] [Accepted: 02/24/2014] [Indexed: 01/01/2023]
Abstract
Homer protein, a member of the post-synaptic density protein family, plays an important role in the neuronal synaptic activity and is extensively involved in neurological disorders. The present study investigates the role of Homer1b/c in modulating neuronal survival by using an in vitro traumatic neuronal injury model, which was achieved by using a punch device that consisted of 28 stainless steel blades joined together and produced 28 parallel cuts. Downregulation of Homer1b/c by specific siRNA significantly (p<0.05) alleviated the cytoplasmic calcium levels and neuron lactate dehydrogenase release, and ultimately decreased the apoptotic rate after traumatic neuronal injury compared with non-targeting siRNA control treatment in cultured rat cortical neurons. Moreover, the expression of metabotropic glutamate receptor 1a (mGluR1a) was significantly (p<0.05) reduced in the Homer1b/c siRNA-transfected neurons after injury. Therefore, Homer1b/c not only modulated the mGluR1a-inositol 1,4,5-triphosphate receptors-Ca(2+) signal transduction pathway, but also regulated the expression of mGluR1a in mechanical neuronal injury. These findings indicate that the suppression of Homer1b/c expression potentially protects neurons from glutamate excitotoxicity after injury and might be an effective intervention target in traumatic brain injury.
Collapse
Affiliation(s)
- F Fei
- Department of Cell Biology, College of Basic Medicine, Fourth Military Medical University, Xi'an 710032, PR China
| | - W Rao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - L Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - B-G Chen
- Central Laboratory, Tongji University Affiliated Shanghai East Hospital, Shanghai 200120, PR China
| | - J Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - Z Fei
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China.
| | - Z Chen
- Department of Cell Biology, College of Basic Medicine, Fourth Military Medical University, Xi'an 710032, PR China.
| |
Collapse
|
11
|
Su JJ, Pan H, Zhou HG, Tang YP, Dong Q, Liu JR. Acid-sensing ion channels activation and hypoxia upregulate Homer1a expression. CNS Neurosci Ther 2014; 20:264-74. [PMID: 24433527 DOI: 10.1111/cns.12206] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 10/18/2013] [Accepted: 10/19/2013] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Recent studies have indicated that dynamic alterations in the structure of postsynaptic density (PSD) are involved in the pathogenesis of many central nervous system disorders, including ischemic stroke. Homer is the newly identified scaffolding protein located at PSD and regulates synaptic function. Homer1a, an immediate early gene, has been shown to be induced by several stimulations, such as glutamate, brain-derived neurotrophic factor, and trauma. However, whether acidosis mediated by acid-sensing ion channels (ASICs) and hypoxia during cerebral ischemia can change Homer1a expression remains to be determined. RESULTS We investigated that acidosis and hypoxia selectively and rapidly upregulated Homer1a expression, but not Homer1b/c in cultured cortical neurons. We also found that Homer1a exhibited induction expression in brain cortex of the middle cerebral artery occlusion (MCAO) rats. Additionally, acid-evoked Homer1a mRNA induction depended on extracellular signal-regulated kinase1/2 (ERK1/2) and Akt activity, and ASIC1a-mediated calcium influx whereas hypoxia depended only on ERK1/2 activity. Also, we demonstrated that continuous acidosis and hypoxia resulted in pronounced cell injury and Homer1a knockdown with small interfering RNA aggravated this damage induced by 3 h acid and hypoxia incubation in neuro-2a cells. CONCLUSION Homer1a might act as an activity-dependent regulator responding to extracellular stimuli during cerebral ischemia.
Collapse
Affiliation(s)
- Jing-Jing Su
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
12
|
Protective effect of Homer 1a on tumor necrosis factor-α with cycloheximide-induced apoptosis is mediated by mitogen-activated protein kinase pathways. Apoptosis 2013; 17:975-88. [PMID: 22660975 DOI: 10.1007/s10495-012-0736-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Although Homer 1, of the postsynaptic density, regulates apoptosis, the signaling mechanisms are not fully elucidated. In this study, we found that tumor necrosis factor-α (TNF-α)/cycloheximide (CHX) treatment transiently increased Homer 1a (the short variant of Homer 1), but did not affect Homer 1b/c (the long variant of Homer 1). Overexpression of Homer 1a blocked TNF-α/CHX-induced apoptotic cell death, whereas inhibition of Homer 1a induction enhanced the pro-apoptotic effect of TNF-α/CHX treatment. Moreover, brain-derived neurotrophic factor, as a potential activator of endogenous Homer 1a, inhibited apoptotic cell death after TNF-α/CHX treatment through induction of Homer 1a. Since three major mitogen-activated protein kinase (MAPK) pathways have important roles in apoptosis, we examined if Homer 1a is involved in the effects of MAPK pathways on apoptosis. It was shown that inhibition of the ERK1/2 pathway increased the expression and the protective effect of Homer 1a, but inhibition of the p38 pathway produced the opposite effect. Cross-talk among MAPK pathways was also associated with the regulation of Homer 1a during apoptotic cell death. Blocking the p38 pathway increased the activity in the ERK1/2 pathway, while inhibition of ERK1/2 pathway abolished the effect of p38 inhibitor on Homer 1a. Furthermore, Homer 1a reversely affected the activation of MAPK pathways. These findings suggest that Homer 1a plays an important role in the prevention of apoptotic cell death and contributes to distinct regulatory effects of MAPK pathways on apoptotic cell death.
Collapse
|
13
|
de Bartolomeis A, Sarappa C, Buonaguro EF, Marmo F, Eramo A, Tomasetti C, Iasevoli F. Different effects of the NMDA receptor antagonists ketamine, MK-801, and memantine on postsynaptic density transcripts and their topography: role of Homer signaling, and implications for novel antipsychotic and pro-cognitive targets in psychosis. Prog Neuropsychopharmacol Biol Psychiatry 2013; 46:1-12. [PMID: 23800465 DOI: 10.1016/j.pnpbp.2013.06.010] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 06/10/2013] [Accepted: 06/14/2013] [Indexed: 12/15/2022]
Abstract
Administration of NMDA receptor antagonists, such as ketamine and MK-801, may induce psychotic-like behaviors in preclinical models of schizophrenia. Ketamine has also been observed to exacerbate psychotic symptoms in schizophrenia patients. However, memantine, a non-competitive NMDA receptor antagonist approved for Alzheimer's disease and proposed for antipsychotic augmentation, may challenge this view. To date, the molecular mechanisms by which these NMDA receptor antagonists cause different neurochemical, behavioral, and clinical effects are still a matter of debate. Here, we investigated by molecular imaging whether these agents could differently modulate gene expression and topographical distribution of glutamatergic postsynaptic density (PSD) proteins. We focused on Homer1a/Homer1b/PSD-95 signaling network, which may be implicated in glutamate-dependent synaptic plasticity, as well as in psychosis pathophysiology and treatment. Ketamine (25 and 50mg/kg) and MK-801 (0.8mg/kg) significantly induced the transcripts of immediate-early genes (Arc, c-fos, and Homer1a) in cortical regions compared to vehicle, whereas they reduced Homer1b and PSD-95 expression in cortical and striatal regions. Differently, memantine (5mg/kg) did not increase Homer1a signal compared to vehicle, whereas it induced c-fos in the somatosensory and in the medial agranular cortices. Moreover, memantine did not affect Homer1b and PSD-95 expression. When compared to ketamine and MK-801, memantine significantly increased the expression of c-fos, Homer1b and PSD-95. Overall, ketamine and MK-801 prominently increased Homer1a/Homer1b expression ratio, whereas memantine elicited the opposite effect. These data may support the view that ketamine, MK-801 and memantine exert divergent effects on PSD transcripts, which may contribute to their partially different behavioral and clinical effects.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine "Federico II", Naples, Italy.
| | | | | | | | | | | | | |
Collapse
|
14
|
Luo P, Li X, Fei Z, Poon W. Scaffold protein Homer 1: implications for neurological diseases. Neurochem Int 2012; 61:731-8. [PMID: 22749857 DOI: 10.1016/j.neuint.2012.06.014] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Revised: 06/16/2012] [Accepted: 06/20/2012] [Indexed: 11/17/2022]
Abstract
Homer proteins are commonly known as scaffold proteins at postsynaptic density. Homer 1 is a widely studied member of the Homer protein family, comprising both synaptic structure and mediating postsynaptic signaling transduction. Both an immediate-early gene encoding a Homer 1 variant and a constitutively expressed Homer 1 variant regulate receptor clustering and trafficking, intracellular calcium homeostasis, and intracellular molecule complex formation. Substantial preclinical investigations have implicated that each of these Homer 1 variants are associated with the etiology of many neurological diseases, such as pain, mental retardation syndromes, Alzheimer's disease, schizophrenia, drug-induced addiction, and traumatic brain injury.
Collapse
Affiliation(s)
- Peng Luo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | | | | | | |
Collapse
|
15
|
Luo P, Chen T, Zhao Y, Xu H, Huo K, Zhao M, Yang Y, Fei Z. Protective effect of Homer 1a against hydrogen peroxide-induced oxidative stress in PC12 cells. Free Radic Res 2012; 46:766-76. [PMID: 22435683 DOI: 10.3109/10715762.2012.678340] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Oxidative stress-induced cell damage is involved in many neurological diseases. Homer protein, as an important scaffold protein at postsynaptic density, regulates synaptic structure and function. Here, we reported that hydrogen peroxide (H(2)O(2)) induced the expression of Homer 1a. Down-regulation of Homer 1a with a specific small interfering RNA (siRNA) exacerbated H(2)O(2)-induced cell injury. Up-regulation of Homer 1a by lentivirus transfection did not affect the anti-oxidant activity, but significantly reduced the reactive oxygen species (ROS) production and lipid peroxidation after H(2)O(2)-induced oxidative stress. Overexpression of Homer 1a attenuated the loss of mitochondrial membrane potential (MMP) and ATP production induced by H(2)O(2), and subsequently inhibited mitochondrial dysfunction-induced cytochrome c release, increase of Bax/Bcl-2 ratio and caspase-9/caspase-3 activity. Furthermore, in the presence of BAPTA-AM, an intracellular free-calcium (Ca(2+)) chelator, overexpression of Homer 1a had no significant effects on H(2)O(2)-induced oxidative stress. These results suggest that Homer 1a has protective effects against H(2)O(2)-induced oxidative stress by reducing ROS accumulation and activation of mitochondrial apoptotic pathway, and these protective effects are dependent on the regulation of intracellular Ca(2+) homeostasis.
Collapse
Affiliation(s)
- Peng Luo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Wang Y, Fei Z, Ma YH, Liu WB, Zhu J, Zhang C, Lin W, Qu Y. VEGF upregulates Homer 1a gene expression via the mitogen-activated protein kinase cascade in cultured cortex neurons. Neurosci Lett 2012; 515:44-9. [DOI: 10.1016/j.neulet.2012.03.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Revised: 03/07/2012] [Accepted: 03/08/2012] [Indexed: 01/21/2023]
|
17
|
Protective effects of mGluR5 positive modulators against traumatic neuronal injury through PKC-dependent activation of MEK/ERK pathway. Neurochem Res 2012; 37:983-90. [PMID: 22228200 DOI: 10.1007/s11064-011-0691-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2011] [Revised: 12/24/2011] [Accepted: 12/29/2011] [Indexed: 12/11/2022]
Abstract
Several previous studies utilizing selective pharmacological antagonists have demonstrated that type 5 metabotropic glutamate receptors (mGluR5) are potential therapeutic targets for the treatment of numerous disorders of the central nervous system, but the role of mGluR5 activation in traumatic brain injury (TBI) is not fully understood. Here in an in vitro TBI model, the mGluR5 agonist (RS)-2-chloro-5- hydroxyphenylglycine (CHPG) and the positive allosteric modulators 3-cyano-N-(1,3- diphenyl-1H-pyrazol-5-yl) benzamide (CDPPB) were used to investigate the neuroprotective potency of mGluR5 activation. Data showed that CHPG and CDPPB suppressed the increase of LDH release and caspase-3 activation induced by traumatic neuronal injury in a dose-dependent manner, and the salutary effects were also present when these compounds were added 1 h after injury. Western blot was used to examine the activation of three members of mitogen-activated protein kinases: extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38 kinase (p38). CHPG and CDPPB enhanced the activation of ERK after traumatic neuronal injury, and PD98059 and U0126, two selective MEK/ERK inhibitors, partly revised the protective effects. Furthermore, we also investigated the role of protein kinase C (PKC) in CHPG and CDPPB-induced neuroprotection. With the pretreatment of chelerythrine chloride, a PKC inhibitor, the surpressing effects of CHPG and CDPPB on traumatic injury-evoked LDH release and caspase-3 activation were blocked. All of these findings extended the protective role of mGluR5 activation in an in vitro model of TBI and suggested that these protective effects might be mediated by the PKC-dependent activation of MEK/ERK pathway. These results may have important implications for the development of mGluR5 modulators to treat TBI.
Collapse
|
18
|
Chen T, Zhang L, Qu Y, Huo K, Jiang X, Fei Z. The selective mGluR5 agonist CHPG protects against traumatic brain injury in vitro and in vivo via ERK and Akt pathway. Int J Mol Med 2011; 29:630-6. [PMID: 22211238 PMCID: PMC3577346 DOI: 10.3892/ijmm.2011.870] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 10/27/2011] [Indexed: 12/31/2022] Open
Abstract
Group I metabotropic glutamate receptors (mGluRs) have been implicated in the pathophysiology of central nervous system injury, but the role of mGluR5 in traumatic brain injury (TBI) remains unclear. In the present study, we investigated the neuroprotective potency of (R,S)-2-chloro-5-hydroxyphenylglycine (CHPG), a selective mGluR5 agonist, for protecting against TBI in both in vitro and in vivo models. Primary cortical neurons were treated with 1 mM CHPG in an in vitro preparation 30 min before TBI, and 250 nM CHPG was injected into the right lateral ventricle of rats 30 min before TBI was induced in in vivo studies. The results showed that CHPG significantly attenuated lactate dehydrogenase (LDH) release and neuronal apoptosis and reduced lesion volume. Compared to the control or vehicle group, the phosphorylation levels of extracellular signal-regulated kinase (ERK) and Akt were increased in the presence of CHPG, even following the induction of TBI. Furthermore, treatment with either the ERK inhibitor PD98059 or Akt inhibitor LY294002 partially reversed the CHPG's neuroprotective effects. These data suggest that CHPG minimizes brain damage after induction of TBI both in vitro and in vivo, and that these protective effects were possibly mediated by activation of the ERK and Akt signaling pathways. Thus, potentiating mGluR5 activity with selective agonists such as CHPG may be useful for the treatment of traumatic brain injury.
Collapse
Affiliation(s)
- Tao Chen
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi 710032, PR China
| | | | | | | | | | | |
Collapse
|
19
|
Luo P, Fei F, Zhang L, Qu Y, Fei Z. The role of glutamate receptors in traumatic brain injury: implications for postsynaptic density in pathophysiology. Brain Res Bull 2011; 85:313-20. [PMID: 21605633 DOI: 10.1016/j.brainresbull.2011.05.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Revised: 04/27/2011] [Accepted: 05/08/2011] [Indexed: 11/25/2022]
Abstract
Traumatic brain injury (TBI) is the major cause of death and disability, and the incidence of TBI continues to increase rapidly. In recent years, increasing attention has been paid to an important structure at the postsynaptic membrane: the postsynaptic density (PSD). Glutamate receptors, as major components of the PSD, are highly responsive to alterations in the glutamate concentration at excitatory synapses and activate intracellular signal transduction via calcium and other second messengers following TBI. PSD scaffold proteins (PSD-95, Homer, and Shank), which anchor glutamate receptors and form a network structure, also have potential effects on these downstream signaling pathways. The changes in the function and structure of these major PSD proteins are also induced by TBI, indicating that there is a more complicated mechanism associated with PSD proteins in the pathophysiological process of TBI.
Collapse
Affiliation(s)
- Peng Luo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, 15 Changle Xi Road, Xi'an 710032, PR China
| | | | | | | | | |
Collapse
|
20
|
Guo WG, Su FF, Yuan LJ, Yang GD, Shi XQ, Li RY, Shu Q, Liu XT, Lu ZF, Zheng QS. Simvastatin inhibits angiotensin II-induced cardiac cell hypertrophy: role of Homer 1a. Clin Exp Pharmacol Physiol 2009; 37:40-5. [PMID: 19515066 DOI: 10.1111/j.1440-1681.2009.05221.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
1. The scaffolding protein Homer 1a is constitutively expressed in the myocardium, although its function in cardiomyocytes remains poorly understood. The aim of the present study was to investigate Homer 1a expression in hypertrophic cardiac cells and its role in angiotensin (Ang) II-induced cardiac hypertrophy. 2. After serum starvation for 24 h, cells were treated with 1 micromol/L simvastatin, 100 nmol/L angiotensin (Ang) II or their combination added to Dulbecco's modified Eagle's medium containing 0.5% serum. For combination treatment with AngII plus simvastatin, cells were exposed to simvastatin 12 h before the addition of AngII to the medium and cells were then incubated in the presence of both drugs for a further 24 h. Western blotting was used to determine Homer 1a protein expression. Hypertrophy was evaluated by determining the protein content per cell. 3. Homer 1a protein levels were upregulated following AngII-induced hypertrophy in H9C2 cells and neonatal rat cardiomyocytes, and these increases were augmented by simvastatin pretreatment. Concomitantly, simvastatin pretreatment inhibited extracellular signal-regulated kinase (ERK) 1/2 phosphorylation and AngII-induced hypertrophy. 4. The inhibitory effects of simvastatin against AngII-induced hypertrophy were attenuated by Homer 1a silencing, suggesting that simvastatin suppresses cardiac hypertrophy in a Homer 1a-dependent manner. Furthermore, AngII-induced hypertrophy and ERK1/2 phosphorylation in neonatal rat cardiomyocytes were significantly inhibited following the overexpression of Homer 1a using an adenovirus. 5. These results suggest a possible role for Homer 1a in inhibiting cardiac hypertrophy perhaps in part through inhibition of ERK1/2 activation.
Collapse
Affiliation(s)
- Wan-Gang Guo
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Fei Z, Zhang X, Bai HM, Jiang XF, Li X, Zhang W, Hu W. Posttraumatic secondary brain insults exacerbates neuronal injury by altering metabotropic glutamate receptors. BMC Neurosci 2007; 8:96. [PMID: 18021417 PMCID: PMC2242800 DOI: 10.1186/1471-2202-8-96] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2007] [Accepted: 11/17/2007] [Indexed: 11/17/2022] Open
Abstract
Background Our previous studies indicated that metabotropic glutamate receptors (mGluRs) are deeply involved in the secondary processes after diffuse brain injury (DBI). In the present study, we used a rodent DBI model to determine whether hypotension exacerbates neuronal injury as a secondary brain insult (SBI) after traumatic brain injury (TBI) by changing the expression of metabotropic glutamate receptors (mGluRs) in the cerebral cortex. Results Three hundred and eleven male Sprague-Dawley rats were randomly assigned into five groups: normal control, sham-operated control, SBI alone, DBI alone, or DBI with SBI. DBI was produced in rats by Marmarou's methods and the SBI model was produced by hypotension. The alteration of neuronal expression of mGluRs after DBI and DBI coupled with SBI was observed by hybridization in situ at different time points in the experiment. We found a higher mortality and neurological severity score (NSS) for rats in the DBI with SBI group compared with those in the DBI alone group. Although there was a significant rise in the expression of group I and group III mGluRs (except mGluR6) and a decrease in the expression of group II mGluRs after DBI (P < 0.05), the changes were more severe when DBI was coupled with SBI (P < 0.05). The expression of group I mGluRs peaked at 24 hours, while the expression of the group III mGluRs peaked at 6 hours after injuries, which may reflect a self-protection first mechanism of the damaged neurons. Moreover, the overall neuro-harmful effects of mGluRs on neurons were seemly associated with higher mortality and NSS in the DBI with SBI group. Conclusion The results suggest posttraumatic SBI may exacerbate neuronal injury or brain injury by altering expression of mGluRs, and more emphasis should therefore be put on the prevention and treatment of SBI.
Collapse
Affiliation(s)
- Zhou Fei
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, PR China.
| | | | | | | | | | | | | |
Collapse
|
22
|
Connell BJ, Crosby KM, Richard MJP, Mayne MB, Saleh TM. Estrogen-mediated neuroprotection in the cortex may require NMDA receptor activation. Neuroscience 2007; 146:160-9. [PMID: 17317017 DOI: 10.1016/j.neuroscience.2007.01.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2006] [Revised: 12/19/2006] [Accepted: 01/11/2007] [Indexed: 11/28/2022]
Abstract
Several studies have suggested that a potential mechanism for estrogen-mediated neuroprotection following experimental stroke is a result of modulating glutamate-mediated excitotoxicity. Our laboratory has shown that in male rats, estrogen injection (systemic or direct intracortical injection) resulted in an immediate depolarization of cortical neurons. Therefore, the present study was designed to investigate whether the estrogen-induced depolarization of cortical neurons was required in mediating the early events associated with this neuroprotection. We tested this hypothesis by co-injecting selective antagonists of the NMDA (MK-801) or AMPA (DNQX) glutamatergic receptors with estrogen. Systemic injection of estrogen significantly attenuated the MK-801-induced decrease in infarct volume following middle cerebral artery occlusion (MCAO). Similarly, when estrogen and MK-801 were co-injected directly into the cortex, no neuroprotection was observed. However, when estrogen or MK-801 was injected centrally 10 min prior to the injection of the other drug, significant neuroprotection was observed. This led us to hypothesize that estrogen-mediated neuroprotection required an initial activation of NMDA receptors. Furthermore, our results suggest that this estrogen-mediated neuroprotection was also associated with a significant increase in m-calpain and activation of an endoplasmic reticulum (ER) specific caspase-12. Finally, the results of current clamp experiments showed that estrogen significantly depolarized cortical neurons as well as enhanced NMDA-induced depolarization. Taken together, these results suggest that estrogen pretreatment may activate NMDA receptors resulting in modification of ER-associated molecular mechanisms involved in neuroprotection following MCAO.
Collapse
Affiliation(s)
- B J Connell
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, P.E.I., Canada C1A 4P3
| | | | | | | | | |
Collapse
|
23
|
Kammermeier PJ. Surface clustering of metabotropic glutamate receptor 1 induced by long Homer proteins. BMC Neurosci 2006; 7:1. [PMID: 16393337 PMCID: PMC1361788 DOI: 10.1186/1471-2202-7-1] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2005] [Accepted: 01/04/2006] [Indexed: 12/02/2022] Open
Abstract
Background Metabotropic glutamate receptors (mGluRs) regulate neuronal excitability and synaptic strength. The group I mGluRs, mGluR1 and 5, are widespread in the brain and localize to post-synaptic sites. The Homer protein family regulates group I mGluR function and distribution. Constitutively expressed 'long' Homer proteins (Homer 1b, 1c, 2 and 3) induce dendritic localization of group I mGluRs and receptor clustering, either internally or on the plasma membrane. Short Homer proteins (Homer 1a, Ania-3) exhibit regulated expression and act as dominant negatives, producing effects on mGluR distribution and function that oppose those of the long Homer proteins. There remains some controversy over whether long Homer proteins induce receptor internalization by inducing retention in the endoplasmic reticulum, or induce mGluR clustering on the plasma membrane. Further, an exhaustive study of the effects of each long Homer isoform on mGluR distribution has not been published. Results The distribution of a GFP-tagged group I mGluR, mGluR1-GFP, was examined in the absence of Homer proteins and in the presence of several Homer isoforms expressed in sympathetic neurons from the rat superior cervical ganglion (SCG) using total internal reflection fluorescence (TIRF-M) and confocal microscopy. Quantitative analysis of mGluR1-GFP fluorescence using TIRF-M revealed that expression of each long Homer isoform tested (Homer 1b, 1c, 2b and 3) induced a significant degree of surface clustering. Using confocal imaging, Homer-induced mGluR clusters were observed intra-cellularly as well as on the plasma membrane. Further, in approximately 40% of neurons co-expressing mGluR1-GFP and Homer 1b, intracellular inclusions were observed, but plasma membrane clusters were also documented in some Homer 1b coexpressing cells. Conclusion All long Homer proteins examined (Homer 1b, 1c, 2b and 3) induced a significant degree of mGluR1-GFP clustering on the plasma membrane compared to cells expressing mGluR1-GFP alone. Clusters induced by long Homers appeared on the plasma membrane and intracellularly, suggesting that clusters form prior to plasma membrane insertion and/or persist after internalization. Finally, while Homer 1b induced surface clustering of mGluR1 in some cells, under some conditions intracellular retention may occur.
Collapse
Affiliation(s)
- Paul J Kammermeier
- Department of Physiology and Pharmacology, Northeastern Ohio Universities College of Medicine, Rootstown, OH 44272, USA.
| |
Collapse
|