201
|
Neural circular RNAs are derived from synaptic genes and regulated by development and plasticity. Nat Neurosci 2015; 18:603-610. [PMID: 25714049 PMCID: PMC4376664 DOI: 10.1038/nn.3975] [Citation(s) in RCA: 893] [Impact Index Per Article: 89.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 02/10/2015] [Indexed: 12/11/2022]
Abstract
Circular RNAs (circRNAs) have re-emerged as an interesting RNA species. Here, by deep RNA profiling in different mouse tissues, we observed that circRNAs are significantly enriched in brain.and a disproportionate fraction of them is derived from host genes that code for synaptic proteins. Moreover, based on separate profiling of the RNAs localized in neuronal cell bodies and neuropil, on average, circRNAs are more enriched in the neuropil than their host gene mRNA isoforms. Using high resolution in situ hybridization we, for the first time, visualized circRNA punctae in the dendrites of neurons. Consistent with the idea that circRNAs might regulate synaptic function, during development, many circRNAs change their abundance abruptly at a time corresponding to synaptogenesis. In addition, following a homeostatic downscaling of neuronal activity many circRNAs exhibit significant up or down-regulation. Together, our data indicate that brain circRNAs are positioned to respond to and regulate synaptic function.
Collapse
|
202
|
Wu SY, Yu MX, Li XG, Xu SF, Shen J, Sun Z, Zhou X, Chen XZ, Tu JC. Identification of Homer1 as a potential prognostic marker for intrahepatic cholangiocarcinoma. Asian Pac J Cancer Prev 2015; 15:3299-304. [PMID: 24815486 DOI: 10.7314/apjcp.2014.15.7.3299] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The aim of the present study was to analyze whether Homer1 is a potential prognostic marker for intrahepatic cholangiocarcinoma (ICC). MATERIALS AND METHODS The expression of Homer1 in ICC tissue was detected with immunohistochemistry and levels of protein in ICC and paratumor tissues were evaluated by Western blotting. Survival analysis by the Kaplan-Meier method was performed to assess prognostic significance. RESULTS Homer1 expression was high in 67.4% (58/86) of ICC samples, and there was significant difference between ICC and adjacent noncancerous tissues (p<0.001); high expression was associated with poor histologic differentiation (p=0.019), TNM stage (p=0.014), lymph node metastasis (p=0.040), and lymphatic invasion (p=0.025). On Kaplan-Meier analysis, a comparison of survival curves of low versus high expressors of Homer1 revealed a highly significant difference in OS (p=0.001) and DFS (p=0.006), indicating that high expression of Homer1 was linked with a worse prognosis. Multivariate analyses showed that Homer1 expression was an independent risk factor predicting overall survival[Hazard ratio(HR), 7.52; 95% confidence interval (CI), 2.63- 21.47; p=0.002] and disease-free survival (HR, 11.56; 95%CI, 5.17-25.96; p<0.001) in ICC. CONCLUSIONS Homer1 promotes lymphatic invasion and associates with lymph node metastasis and poor prognosis of ICC. The current study shows that Homer1 may be an independent prognostic factor for ICC patients after curative resection, and it provides an important basis for screening/treating high-risk patients.
Collapse
Affiliation(s)
- San-Yun Wu
- Department of Clinical Laboratory Medicine and Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, China E-mail :
| | | | | | | | | | | | | | | | | |
Collapse
|
203
|
Gould AT, Sacramento AD, Wroten MG, Miller BW, von Jonquieres G, Klugmann M, Ben-Shahar O, Szumlinski KK. Cocaine-elicited imbalances in ventromedial prefrontal cortex Homer1 versus Homer2 expression: implications for relapse. Addict Biol 2015; 20:148-57. [PMID: 24118426 DOI: 10.1111/adb.12088] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Withdrawal from a history of extended access to self-administered cocaine produces a time-dependent intensification of drug seeking, which might relate to a cocaine-induced imbalance in the relative expression of constitutively expressed Homer1 versus Homer2 isoforms within the ventromedial aspect of the prefrontal cortex (vmPFC). Thus, we employed immunoblotting to examine the relation between cue-reinforced lever pressing at 3- versus 30-day withdrawal from a 10-day history of extended access (6 hours/day) to intravenous cocaine (0.25 mg/infusion) or saline (Sal6h), and the expression of Homer1b/c and Homer2a/b within the vmPFC versus the more dorsomedial aspect of this structure (dmPFC). Behavioral studies employed adeno-associated virus (AAV) vectors to reverse cocaine-elicited changes in the relative expression of Homer1 versus Homer2 isoforms and tested animals for cocaine prime-, and cue-induced responding following extinction training. Cocaine self-administration elevated both Homer1b/c and Homer2a/b levels within the vmPFC at 3-day withdrawal, and the rise in Homer2a/b persisted for at least 30 days. dmPFC Homer levels did not change as a function of self-administration history. Reversing the relative increase in Homer2 versus Homer1 expression via Homer1c overexpression or Homer2b knockdown failed to influence cue-reinforced lever pressing when animals were tested in a drug-free state, but both AAV treatments prevented cocaine-primed reinstatement of lever-pressing behavior. These data suggest that a cocaine-elicited imbalance in the relative expression of constitutively expressed Homer2 versus Homer1 within the vmPFC is necessary for the capacity of cocaine to reinstate drug-seeking behavior, posing drug-induced changes in vmPFC Homer expression as a molecular trigger contributing to drug-elicited relapse.
Collapse
Affiliation(s)
- Adam T. Gould
- Department of Psychological and Brain Sciences and the Neuroscience Research Institute; University of California at Santa Barbara; Santa Barbara CA USA
| | - Arianne D. Sacramento
- Department of Psychological and Brain Sciences and the Neuroscience Research Institute; University of California at Santa Barbara; Santa Barbara CA USA
| | - Melissa G. Wroten
- Department of Psychological and Brain Sciences and the Neuroscience Research Institute; University of California at Santa Barbara; Santa Barbara CA USA
| | - Bailey W. Miller
- Department of Psychological and Brain Sciences and the Neuroscience Research Institute; University of California at Santa Barbara; Santa Barbara CA USA
| | - Georg von Jonquieres
- Translational Neuroscience Facility; School of Medical Sciences; University of New South Wales; Australia
| | - Matthias Klugmann
- Translational Neuroscience Facility; School of Medical Sciences; University of New South Wales; Australia
| | - Osnat Ben-Shahar
- Department of Psychological and Brain Sciences and the Neuroscience Research Institute; University of California at Santa Barbara; Santa Barbara CA USA
| | - Karen K. Szumlinski
- Department of Psychological and Brain Sciences and the Neuroscience Research Institute; University of California at Santa Barbara; Santa Barbara CA USA
| |
Collapse
|
204
|
Ménard C, Gaudreau P, Quirion R. Signaling pathways relevant to cognition-enhancing drug targets. Handb Exp Pharmacol 2015; 228:59-98. [PMID: 25977080 DOI: 10.1007/978-3-319-16522-6_3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Aging is generally associated with a certain cognitive decline. However, individual differences exist. While age-related memory deficits can be observed in humans and rodents in the absence of pathological conditions, some individuals maintain intact cognitive functions up to an advanced age. The mechanisms underlying learning and memory processes involve the recruitment of multiple signaling pathways and gene expression, leading to adaptative neuronal plasticity and long-lasting changes in brain circuitry. This chapter summarizes the current understanding of how these signaling cascades could be modulated by cognition-enhancing agents favoring memory formation and successful aging. It focuses on data obtained in rodents, particularly in the rat as it is the most common animal model studied in this field. First, we will discuss the role of the excitatory neurotransmitter glutamate and its receptors, downstream signaling effectors [e.g., calcium/calmodulin-dependent protein kinase II (CaMKII), protein kinase C (PKC), extracellular signal-regulated kinases (ERK), mammalian target of rapamycin (mTOR), cAMP response element-binding protein (CREB)], associated immediate early gene (e.g., Homer 1a, Arc and Zif268), and growth factors [insulin-like growth factors (IGFs) and brain-derived neurotrophic factor (BDNF)] in synaptic plasticity and memory formation. Second, the impact of the cholinergic system and related modulators on memory will be briefly reviewed. Finally, since dynorphin neuropeptides have recently been associated with memory impairments in aging, it is proposed as an attractive target to develop novel cognition-enhancing agents.
Collapse
Affiliation(s)
- Caroline Ménard
- Douglas Mental Health University Institute, McGill University, Perry Pavilion, 6875 LaSalle Boulevard, Montreal, QC, Canada, H4H 1R3
| | | | | |
Collapse
|
205
|
Diagnostic potential of differentially expressed Homer1, IL-1β, and TNF-α in coronary artery disease. Int J Mol Sci 2014; 16:535-46. [PMID: 25551602 PMCID: PMC4307261 DOI: 10.3390/ijms16010535] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 12/22/2014] [Indexed: 11/22/2022] Open
Abstract
Increasing evidences suggest that inflammation plays an important role in the pathogenesis of coronary artery disease (CAD). Numerous inflammatory cytokines and related genes mediate adverse cardiovascular events in patients with CAD, such as interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and Homer in the present study. The study was carried out on 163 CAD patients at different stages and 68 controls. The gene expression of Homer1, Homer2, Homer3, IL-1β, and TNF-α in the peripheral blood leukocytes were measured by real-time polymerase chain reaction. The mRNA levels of Homer1, IL-1β, and TNF-α in CAD patients were significantly higher than those in the control group, but not Homer2 and Homer3. However, there was no considerable difference in the mRNA levels of Homer1, IL-1β, and TNF-α among AMI, UAP, and SAP three subgroups of CAD. The receiver operating characteristic (ROC) curves showed that Homer1 had a better diagnostic value for UAP patients compared with IL-1β and TNF-α. Like IL-1β and TNF-α, Homer1 may also be an important participant of atherosclerotic plaque development and eventually rupture. The results of the present study may provide an important basis for diagnosing CAD patients, and provide new therapeutic targets for CAD.
Collapse
|
206
|
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
|
207
|
Crosby ND, Zaucke F, Kras JV, Dong L, Luo ZD, Winkelstein BA. Thrombospondin-4 and excitatory synaptogenesis promote spinal sensitization after painful mechanical joint injury. Exp Neurol 2014; 264:111-20. [PMID: 25483397 DOI: 10.1016/j.expneurol.2014.11.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 11/24/2014] [Accepted: 11/26/2014] [Indexed: 02/08/2023]
Abstract
Facet joint injury induces persistent pain that may be maintained by structural plasticity in the spinal cord. Astrocyte-derived thrombospondins, especially thrombospondin-4 (TSP4), have been implicated in synaptogenesis and spinal sensitization in neuropathic pain, but the TSP4 response and its relationship to synaptic changes in the spinal cord have not been investigated for painful joint injury. This study investigates the role of TSP4 in the development and maintenance of persistent pain following injurious facet joint distraction in rats and tests the hypothesis that excitatory synaptogenesis contributes to such pain. Painful facet joint loading induces dorsal horn excitatory synaptogenesis along with decreased TSP4 in the DRG and increased astrocytic release of TSP4 in the spinal cord, all of which parallel the time course of sustained tactile allodynia. Blocking injury-induced spinal TSP4 expression with antisense oligonucleotides or reducing TSP4 activity at its neuronal receptor in the spinal cord with gabapentin treatment both attenuate the allodynia and dorsal horn synaptogenesis that develop after painful facet joint loading. Increased spinal TSP4 also facilitates the development of allodynia and spinal hyperexcitability, even after non-painful physiological loading of the facet joint. These results suggest that spinal TSP4 plays an important role in the development and maintenance of persistent joint-mediated pain by inducing excitatory synaptogenesis and facilitating the transduction of mechanical loading of the facet joint that leads to spinal hyperexcitability.
Collapse
Affiliation(s)
- Nathan D Crosby
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Frank Zaucke
- Center for Biochemistry, Medical Faculty, University of Cologne, D-50931 Cologne, Germany
| | - Jeffrey V Kras
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Ling Dong
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Z David Luo
- Department of Anesthesiology and Perioperative Care, University of California Irvine Medical Center, Irvine, CA 92868, United States; Department of Pharmacology, University of California Irvine Medical Center, Irvine, CA 92868, United States
| | - Beth A Winkelstein
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, United States; Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
208
|
Dittrich F, Ramenda C, Grillitsch D, Frankl-Vilches C, Ko MC, Hertel M, Goymann W, ter Maat A, Gahr M. Regulatory mechanisms of testosterone-stimulated song in the sensorimotor nucleus HVC of female songbirds. BMC Neurosci 2014; 15:128. [PMID: 25442096 PMCID: PMC4261767 DOI: 10.1186/s12868-014-0128-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 11/13/2014] [Indexed: 01/04/2023] Open
Abstract
Background In male birds, influence of the sex steroid hormone testosterone and its estrogenic metabolites on seasonal song behavior has been demonstrated for many species. In contrast, female song was only recently recognized to be widespread among songbird species, and to date, sex hormone effects on singing and brain regions controlling song development and production (song control nuclei) have been studied in females almost exclusively using domesticated canaries (Serinus canaria). However, domesticated female canaries hardly sing at all in normal circumstances and exhibit only very weak, if any, song seasonally under the natural photoperiod. By contrast, adult female European robins (Erithacus rubecula) routinely sing during the winter season, a time when they defend feeding territories and show elevated circulating testosterone levels. We therefore used wild female European robins captured in the fall to examine the effects of testosterone administration on song as well as on the anatomy and the transcriptome of the song control nucleus HVC (sic). The results obtained from female robins were compared to outcomes of a similar experiment done in female domesticated canaries. Results Testosterone treatment induced abundant song in female robins. Examination of HVC transcriptomes and histological analyses of song control nuclei showed testosterone-induced differentiation processes related to neuron growth and spacing, angiogenesis and neuron projection morphogenesis. Similar effects were found in female canaries treated with testosterone. In contrast, the expression of genes related to synaptic transmission was not enhanced in the HVC of testosterone treated female robins but was strongly up-regulated in female canaries. A comparison of the testosterone-stimulated transcriptomes indicated that brain-derived neurotrophic factor (BDNF) likely functions as a common mediator of the testosterone effects in HVC. Conclusions Testosterone-induced singing of female robins correlated with cellular differentiation processes in the HVC that were partially similar to those seen in the HVC of testosterone-treated female canaries. Other modes of testosterone action, notably related to synaptic transmission, appeared to be regulated in a more species-specific manner in the female HVC. Divergent effects of testosterone on the HVC of different species might be related to differences between species in regulatory mechanisms of the singing behavior. Electronic supplementary material The online version of this article (doi:10.1186/s12868-014-0128-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Falk Dittrich
- Max Planck Institute for Ornithology, Department of Behavioural Neurobiology, Eberhard-Gwinner Strasse, Haus 6a, Seewiesen, 82319, Germany.
| | - Claudia Ramenda
- Max Planck Institute for Ornithology, Department of Behavioural Neurobiology, Eberhard-Gwinner Strasse, Haus 6a, Seewiesen, 82319, Germany.
| | - Doris Grillitsch
- Max Planck Institute for Ornithology, Department of Behavioural Neurobiology, Eberhard-Gwinner Strasse, Haus 6a, Seewiesen, 82319, Germany.
| | - Carolina Frankl-Vilches
- Max Planck Institute for Ornithology, Department of Behavioural Neurobiology, Eberhard-Gwinner Strasse, Haus 6a, Seewiesen, 82319, Germany.
| | - Meng-Ching Ko
- Max Planck Institute for Ornithology, Department of Behavioural Neurobiology, Eberhard-Gwinner Strasse, Haus 6a, Seewiesen, 82319, Germany.
| | - Moritz Hertel
- Max Planck Institute for Ornithology, Department of Behavioural Neurobiology, Eberhard-Gwinner Strasse, Haus 6a, Seewiesen, 82319, Germany.
| | - Wolfgang Goymann
- Max Planck Institute for Ornithology, Department of Behavioural Neurobiology, Eberhard-Gwinner Strasse, Haus 6a, Seewiesen, 82319, Germany.
| | - Andries ter Maat
- Max Planck Institute for Ornithology, Department of Behavioural Neurobiology, Eberhard-Gwinner Strasse, Haus 6a, Seewiesen, 82319, Germany.
| | - Manfred Gahr
- Max Planck Institute for Ornithology, Department of Behavioural Neurobiology, Eberhard-Gwinner Strasse, Haus 6a, Seewiesen, 82319, Germany.
| |
Collapse
|
209
|
Developmental and activity-dependent expression of LanCL1 confers antioxidant activity required for neuronal survival. Dev Cell 2014; 30:479-87. [PMID: 25158856 DOI: 10.1016/j.devcel.2014.06.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 05/09/2014] [Accepted: 06/13/2014] [Indexed: 02/05/2023]
Abstract
Production of reactive oxygen species (ROS) increases with neuronal activity that accompanies synaptic development and function. Transcription-related factors and metabolic enzymes that are expressed in all tissues have been described to counteract neuronal ROS to prevent oxidative damage. Here, we describe the antioxidant gene LanCL1 that is prominently enriched in brain neurons. Its expression is developmentally regulated and induced by neuronal activity, neurotrophic factors implicated in neuronal plasticity and survival, and oxidative stress. Genetic deletion of LanCL1 causes enhanced accumulation of ROS in brain, as well as development-related lipid, protein, and DNA damage; mitochondrial dysfunction; and apoptotic neurodegeneration. LanCL1 transgene protects neurons from ROS. LanCL1 protein purified from eukaryotic cells catalyzes the formation of thioether products similar to glutathione S-transferase. These studies reveal a neuron-specific glutathione defense mechanism that is essential for neuronal function and survival.
Collapse
|
210
|
Mansouri M, Kasugai Y, Fukazawa Y, Bertaso F, Raynaud F, Perroy J, Fagni L, Kaufmann WA, Watanabe M, Shigemoto R, Ferraguti F. Distinct subsynaptic localization of type 1 metabotropic glutamate receptors at glutamatergic and
GABA
ergic synapses in the rodent cerebellar cortex. Eur J Neurosci 2014; 41:157-67. [DOI: 10.1111/ejn.12779] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 09/26/2014] [Accepted: 10/05/2014] [Indexed: 11/30/2022]
Affiliation(s)
- Mahnaz Mansouri
- Department of Pharmacology Innsbruck Medical University Innsbruck Austria
| | - Yu Kasugai
- Department of Pharmacology Innsbruck Medical University Innsbruck Austria
| | - Yugo Fukazawa
- Division of Cerebral Structure National Institute for Physiological Sciences Okazaki Japan
| | - Federica Bertaso
- CNRS UMR‐5203 Institut de Génomique Fonctionnelle Montpellier France
- INSERM U661 Montpellier France
- Universités de Montpellier 1 & 2 UMR‐5203 Montpellier France
| | - Fabrice Raynaud
- CNRS UMR‐5203 Institut de Génomique Fonctionnelle Montpellier France
- INSERM U661 Montpellier France
- Universités de Montpellier 1 & 2 UMR‐5203 Montpellier France
| | - Julie Perroy
- CNRS UMR‐5203 Institut de Génomique Fonctionnelle Montpellier France
- INSERM U661 Montpellier France
- Universités de Montpellier 1 & 2 UMR‐5203 Montpellier France
| | - Laurent Fagni
- CNRS UMR‐5203 Institut de Génomique Fonctionnelle Montpellier France
- INSERM U661 Montpellier France
- Universités de Montpellier 1 & 2 UMR‐5203 Montpellier France
| | - Walter A. Kaufmann
- Department of Pharmacology Innsbruck Medical University Innsbruck Austria
| | | | - Ryuichi Shigemoto
- Division of Cerebral Structure National Institute for Physiological Sciences Okazaki Japan
| | | |
Collapse
|
211
|
Sevastyanova TN, Kammermeier PJ. Cooperative signaling between homodimers of metabotropic glutamate receptors 1 and 5. Mol Pharmacol 2014; 86:492-504. [PMID: 25113912 PMCID: PMC4201138 DOI: 10.1124/mol.114.093468] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 08/11/2014] [Indexed: 11/22/2022] Open
Abstract
Metabotropic glutamate receptors (mGluRs) function as dimers. Recent work suggests that mGluR1 and mGluR5 may physically interact, but the nature and functional consequences of this relationship have not been addressed. In this study, the functional and pharmacological consequences of this interaction were investigated. Using heterologous expression of mGluR cDNA in rat sympathetic neurons from the superior cervical ganglion and inhibition of the native calcium currents as an assay for receptor activation, a functional interdependence between mGluR1 and mGluR5 was demonstrated. In neurons coexpressing these receptors, combining a selective mGluR1 competitive antagonist with either an mGluR1- or mGluR5-selective negative allosteric modulator (NAM) BAY36-7620 [(3aS,6aS)-hexahydro-5-methylene-6a-(2-naphthalenylmethyl)-1H-cyclopenta[c]furan-1-one] or MPEP [2-methyl-6-(phenylethynyl)pyridine hydrochloride], respectively, strongly occluded signaling by both receptors to an approximately equal degree. By contrast, in cells coexpressing mGluR1 and mGluR2, combining the same mGluR1 competitive inhibitor with an mGluR1 or mGluR2 NAM yielded partial and full inhibition of the response, respectively, as expected for independently acting receptors. In neurons expressing mGluR1 and mGluR5, the selective NAMs each strongly inhibited the response to glutamate, suggesting that these receptors do not interact as heterodimers, which would not be inhibited by selective NAMs. Finally, evidence for a similar mGluR1/mGluR5 functional dependence is shown in medium spiny striatal neurons. Together, these data demonstrate cooperative signaling between mGluR1 and mGluR5 in a manner inconsistent with heterodimerization, and thus suggest an interaction between homodimers.
Collapse
Affiliation(s)
- Tatyana N Sevastyanova
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York
| | - Paul J Kammermeier
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
212
|
Fan P, Cunliffe HE, Griffith OL, Agboke FA, Ramos P, Gray JW, Jordan VC. Identification of gene regulation patterns underlying both oestrogen- and tamoxifen-stimulated cell growth through global gene expression profiling in breast cancer cells. Eur J Cancer 2014; 50:2877-86. [PMID: 25212499 PMCID: PMC4210771 DOI: 10.1016/j.ejca.2014.08.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 07/22/2014] [Accepted: 08/07/2014] [Indexed: 01/13/2023]
Abstract
PURPOSE A c-Src inhibitor blocks oestrogen (E2)-induced stress and converts E2 responses from inducing apoptosis to growth stimulation in E2-deprived breast cancer cells. A reprogrammed cell line, MCF-7:PF, results in a functional oestrogen receptor (ER). We addressed the question of whether the selective ER modulator 4-hydroxytamoxifen (4-OHT) could target ER to prevent E2-stimulated growth in MCF-7:PF cells. METHODS Expression of mRNA was measured through real-time RT-PCR. Global gene expression profile was analysed through microarray. Transcriptome profiles were screened by RNA-sequencing. RESULTS Unexpectedly, both 4-OHT and E2 stimulated cell growth in a concentration-dependent manner. Expression profiling showed a remarkable overlap in genes regulated in the same direction by E2 and 4-OHT. Pathway enrichment analysis of the 280 genes commonly deregulated in MCF-7:PF cells by 4-OHT and E2 revealed functions mainly related to membrane, cytoplasm and metabolic processes. Further analysis of 98 genes up-regulated by both 4-OHT and E2 uncovered a significant enrichment in genes associated with membrane remodelling, cytoskeleton reorganisation, cytoplasmic adapter proteins, cytoplasm organelle proteins and related processes. 4-OHT was more potent than E2 in up-regulating some membrane remodelling molecules, such as EHD2, FHL2, HOMER3 and RHOF. In contrast, 4-OHT acted as an antagonist to inhibit expression of the majority of enriched membrane-associated genes in wild-type MCF-7 cells. CONCLUSIONS Long-term selection pressure has changed the cell population responses to 4-OHT. Membrane-associated signalling is critical for 4-OHT-stimulated cell growth in MCF-7:PF cells. This study provides a rationale for the further investigation of target therapy for tamoxifen resistant patients.
Collapse
Affiliation(s)
- Ping Fan
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC 20057, United States
| | - Heather E Cunliffe
- Computational Biology Division, The Translational Genomics Research Institute, Phoenix, AZ 85004, United States
| | - Obi L Griffith
- Department of Medicine, Division of Oncology, The Genome Institute, Washington University, St. Louis, MO 63108, United States
| | - Fadeke A Agboke
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC 20057, United States
| | - Pilar Ramos
- Computational Biology Division, The Translational Genomics Research Institute, Phoenix, AZ 85004, United States
| | - Joe W Gray
- Biomedical Engineering Department, Oregon Health and Science University, Portland, OR 97239, United States
| | - V Craig Jordan
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC 20057, United States.
| |
Collapse
|
213
|
Stress hormone exposure reduces mGluR5 expression in the nucleus accumbens: functional implications for interoceptive sensitivity to alcohol. Neuropsychopharmacology 2014; 39:2376-86. [PMID: 24713611 PMCID: PMC4138747 DOI: 10.1038/npp.2014.85] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 03/11/2014] [Accepted: 04/02/2014] [Indexed: 12/19/2022]
Abstract
Escalations in alcohol drinking associated with experiencing stressful life events and chronic life stressors may be related to altered sensitivity to the interoceptive/subjective effects of alcohol. Indeed, through the use of drug discrimination methods, rats show decreased sensitivity to the discriminative stimulus (interoceptive) effects of alcohol following exposure to the stress hormone corticosterone (CORT). This exposure produces heightened elevations in plasma CORT levels (eg, as may be experienced by an individual during stressful episodes). We hypothesized that decreased sensitivity to alcohol may be related, in part, to changes in metabotropic glutamate receptors-subtype 5 (mGluR5) in the nucleus accumbens, as these receptors in this brain region are known to regulate the discriminative stimulus effects of alcohol. In the accumbens, we found reduced mGluR5 expression (immunohistochemistry and Western blot) and decreased neural activation (as measured by c-Fos immunohistochemistry) in response to a moderate alcohol dose (1 g/kg) following CORT exposure (7 days). The functional role of these CORT-induced adaptations in relation to the discriminative stimulus effects of alcohol was confirmed, as both the systemic administration of 3-Cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB) an mGluR5 positive allosteric modulator and the intra-accumbens administration of (R,S)-2-Amino-2-(2-chloro-5-hydroxyphenyl)acetic acid sodium salt (CHPG) an mGluR5 agonist restored sensitivity to alcohol in discrimination-trained rats. These results suggest that activation of mGluR5 may alleviate the functional impact of the CORT-induced downregulation of mGluR5 in relation to sensitivity to alcohol. Understanding the contribution of such neuroadaptations to the interoceptive effects of alcohol may enrich our understanding of potential changes in subjective sensitivity to alcohol during stressful episodes.
Collapse
|
214
|
Nowak G, Pomierny-Chamioło L, Siwek A, Niedzielska E, Pomierny B, Pałucha-Poniewiera A, Pilc A. Prolonged administration of antidepressant drugs leads to increased binding of [3H]MPEP to mGlu5 receptors. Neuropharmacology 2014; 84:46-51. [DOI: 10.1016/j.neuropharm.2014.04.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 04/17/2014] [Accepted: 04/22/2014] [Indexed: 11/28/2022]
|
215
|
Yang YM, Lee J, Jo H, Park S, Chang I, Muallem S, Shin DM. Homer2 protein regulates plasma membrane Ca²⁺-ATPase-mediated Ca²⁺ signaling in mouse parotid gland acinar cells. J Biol Chem 2014; 289:24971-9. [PMID: 25049230 DOI: 10.1074/jbc.m114.577221] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Homer proteins are scaffold molecules with a domain structure consisting of an N-terminal Ena/VASP homology 1 protein-binding domain and a C-terminal leucine zipper/coiled-coil domain. The Ena/VASP homology 1 domain recognizes proline-rich motifs and binds multiple Ca(2+)-signaling proteins, including G protein-coupled receptors, inositol 1,4,5-triphosphate receptors, ryanodine receptors, and transient receptor potential channels. However, their role in Ca(2+) signaling in nonexcitable cells is not well understood. In this study, we investigated the role of Homer2 on Ca(2+) signaling in parotid gland acinar cells using Homer2-deficient (Homer2(-/-)) mice. Homer2 is localized at the apical pole in acinar cells. Deletion of Homer2 did not affect inositol 1,4,5-triphosphate receptor localization or channel activity and did not affect the expression and activity of sarco/endoplasmic reticulum Ca(2+)-ATPase pumps. In contrast, Homer2 deletion markedly increased expression of plasma membrane Ca(2+)-ATPase (PMCA) pumps, in particular PMCA4, at the apical pole. Accordingly, Homer2 deficiency increased Ca(2+) extrusion by acinar cells. These findings were supported by co-immunoprecipitation of Homer2 and PMCA in wild-type parotid cells and transfected human embryonic kidney 293 (HEK293) cells. We identified a Homer-binding PPXXF-like motif in the N terminus of PMCA that is required for interaction with Homer2. Mutation of the PPXXF-like motif did not affect the interaction of PMCA with Homer1 but inhibited its interaction with Homer2 and increased Ca(2+) clearance by PMCA. These findings reveal an important regulation of PMCA by Homer2 that has a central role on PMCA-mediated Ca(2+) signaling in parotid acinar cells.
Collapse
Affiliation(s)
- Yu-Mi Yang
- From the Department of Oral Biology, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul 120-752, Korea
| | - Jiae Lee
- From the Department of Oral Biology, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul 120-752, Korea
| | - Hae Jo
- the College of Life Sciences and Graduate School of Biotechnology, Kyunghee University, Global Campus, Gyeonggi 446-701, Korea, and
| | - Soonhong Park
- From the Department of Oral Biology, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul 120-752, Korea
| | - Inik Chang
- From the Department of Oral Biology, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul 120-752, Korea
| | - Shmuel Muallem
- the Epithelial Signaling and Transport Section, Molecular Physiology and Therapeutics Branch, NIDCR, National Institutes of Health, Bethesda, Maryland 20892
| | - Dong Min Shin
- From the Department of Oral Biology, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul 120-752, Korea,
| |
Collapse
|
216
|
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
|
217
|
Burkhardt P, Grønborg M, McDonald K, Sulur T, Wang Q, King N. Evolutionary insights into premetazoan functions of the neuronal protein homer. Mol Biol Evol 2014; 31:2342-55. [PMID: 24899667 PMCID: PMC4137706 DOI: 10.1093/molbev/msu178] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Reconstructing the evolution and ancestral functions of synaptic proteins promises to shed light on how neurons first evolved. The postsynaptic density (PSD) protein Homer scaffolds membrane receptors and regulates Ca2+ signaling in diverse metazoan cell types (including neurons and muscle cells), yet its ancestry and core functions are poorly understood. We find that the protein domain organization and essential biochemical properties of metazoan Homer proteins, including their ability to tetramerize, are conserved in the choanoflagellate Salpingoeca rosetta, one of the closest living relatives of metazoans. Unlike in neurons, Homer localizes to the nucleoplasm in S. rosetta and interacts directly with Flotillin, a protein more commonly associated with cell membranes. Surprisingly, we found that the Homer/Flotillin interaction and its localization to the nucleus are conserved in metazoan astrocytes. These findings suggest that Homer originally interacted with Flotillin in the nucleus of the last common ancestor of metazoans and choanoflagellates and was later co-opted to function as a membrane receptor scaffold in the PSD.
Collapse
Affiliation(s)
- Pawel Burkhardt
- Howard Hughes Medical Institute and the Department of Molecular and Cell Biology, University of California, Berkeley
| | | | - Kent McDonald
- Electron Microscopy Laboratory, University of California, Berkeley
| | - Tara Sulur
- Howard Hughes Medical Institute and the Department of Molecular and Cell Biology, University of California, Berkeley
| | - Qi Wang
- California Institute for Quantitative Biosciences, University of California, Berkeley
| | - Nicole King
- Howard Hughes Medical Institute and the Department of Molecular and Cell Biology, University of California, Berkeley
| |
Collapse
|
218
|
de Bartolomeis A, Buonaguro EF, Iasevoli F, Tomasetti C. The emerging role of dopamine-glutamate interaction and of the postsynaptic density in bipolar disorder pathophysiology: Implications for treatment. J Psychopharmacol 2014; 28:505-26. [PMID: 24554693 DOI: 10.1177/0269881114523864] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Aberrant synaptic plasticity, originating from abnormalities in dopamine and/or glutamate transduction pathways, may contribute to the complex clinical manifestations of bipolar disorder (BD). Dopamine and glutamate systems cross-talk at multiple levels, such as at the postsynaptic density (PSD). The PSD is a structural and functional protein mesh implicated in dopamine and glutamate-mediated synaptic plasticity. Proteins at PSD have been demonstrated to be involved in mood disorders pathophysiology and to be modulated by antipsychotics and mood stabilizers. On the other side, post-receptor effectors such as protein kinase B (Akt), glycogen synthase kinase-3 (GSK-3) and the extracellular signal-regulated kinase (Erk), which are implicated in both molecular abnormalities and treatment of BD, may interact with PSD proteins, and participate in the interplay of the dopamine-glutamate signalling pathway. In this review, we describe emerging evidence on the molecular cross-talk between dopamine and glutamate signalling in BD pathophysiology and pharmacological treatment, mainly focusing on dysfunctions in PSD molecules. We also aim to discuss future therapeutic strategies that could selectively target the PSD-mediated signalling cascade at the crossroads of dopamine-glutamate neurotransmission.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Section of Psychiatry, University Medical School of Naples "Federico II", Naples, Italy
| | - Elisabetta F Buonaguro
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Section of Psychiatry, University Medical School of Naples "Federico II", Naples, Italy
| | - Felice Iasevoli
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Section of Psychiatry, University Medical School of Naples "Federico II", Naples, Italy
| | - Carmine Tomasetti
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Section of Psychiatry, University Medical School of Naples "Federico II", Naples, Italy
| |
Collapse
|
219
|
Changes in mGlu5 receptor-dependent synaptic plasticity and coupling to homer proteins in the hippocampus of Ube3A hemizygous mice modeling angelman syndrome. J Neurosci 2014; 34:4558-66. [PMID: 24672001 DOI: 10.1523/jneurosci.1846-13.2014] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Angelman syndrome (AS) is caused by the loss of Ube3A, an ubiquitin ligase that commits specific proteins to proteasomal degradation. How this defect causes autism and other pathological phenotypes associated with AS is unknown. Long-term depression (LTD) of excitatory synaptic transmission mediated by type 5 metabotropic glutamate (mGlu5) receptors was enhanced in hippocampal slices of Ube3A(m-/p+) mice, which model AS. No changes were found in NMDA-dependent LTD induced by low-frequency stimulation. mGlu5 receptor-dependent LTD in AS mice was sensitive to the protein synthesis inhibitor anisomycin, and relied on the same signaling pathways as in wild-type mice, e.g., the mitogen-activated protein kinase (MAPK) pathway, the phosphatidylinositol-3-kinase (PI3K)/mammalian target of rapamycine pathway, and protein tyrosine phosphatase. Neither the stimulation of MAPK and PI3K nor the increase in Arc (activity-regulated cytoskeleton-associated protein) levels in response to mGlu5 receptor activation were abnormal in hippocampal slices from AS mice compared with wild-type mice. mGlu5 receptor expression and mGlu1/5 receptor-mediated polyphosphoinositide hydrolysis were also unchanged in the hippocampus of AS mice. In contrast, AS mice showed a reduced expression of the short Homer protein isoform Homer 1a, and an increased coupling of mGlu5 receptors to Homer 1b/c proteins in the hippocampus. These findings support the link between Homer proteins and monogenic autism, and lay the groundwork for the use of mGlu5 receptor antagonists in AS.
Collapse
|
220
|
Luo P, Chen T, Zhao Y, Zhang L, Yang Y, Liu W, Li S, Rao W, Dai S, Yang J, Fei Z. Postsynaptic scaffold protein Homer 1a protects against traumatic brain injury via regulating group I metabotropic glutamate receptors. Cell Death Dis 2014; 5:e1174. [PMID: 24722299 PMCID: PMC5424101 DOI: 10.1038/cddis.2014.116] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Revised: 02/11/2014] [Accepted: 02/19/2014] [Indexed: 12/22/2022]
Abstract
Traumatic brain injury (TBI) produces excessive glutamate, leading to excitotoxicity via the activation of glutamate receptors. Postsynaptic density scaffold proteins have crucial roles in mediating signal transduction from glutamate receptors to their downstream mediators. Therefore, studies on the mechanisms underlying regulation of excitotoxicity by scaffold proteins can uncover new treatments for TBI. Here, we demonstrated that the postsynaptic scaffold protein Homer 1a was neuroprotective against TBI in vitro and in vivo, and this neuroprotection was associated with its effects on group I metabotropic glutamate receptors (mGluRs). Upon further study, we found that Homer 1a mainly affected neuronal injury induced by mGluR1 activation after TBI and also influenced mGluR5 function when its activity was restored. The ability of Homer 1a to disrupt mGluR-ERK signaling contributed to its ability to regulate the functions of mGluR1 and mGluR5 after traumatic injury. Intracellular Ca(2+) and PKC were two important factors involved in the mediation of mGluR-ERK signaling by Homer 1a. These results define Homer 1a as a novel endogenous neuroprotective agent against TBI.
Collapse
Affiliation(s)
- P Luo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - T Chen
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Y Zhao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - L Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Y Yang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - W Liu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - S Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - W Rao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - S Dai
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - J Yang
- Department of Surgery, Boston Veterans Affairs Healthcare System, Boston University School of Medicine, Boston, MA, USA
| | - Z Fei
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| |
Collapse
|
221
|
The synaptic targeting of mGluR1 by its carboxyl-terminal domain is crucial for cerebellar function. J Neurosci 2014; 34:2702-12. [PMID: 24523559 DOI: 10.1523/jneurosci.3542-13.2014] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The metabotropic glutamate receptor subtype 1 (mGluR1, Grm1) in cerebellar Purkinje cells (PCs) is essential for motor coordination and motor learning. At the synaptic level, mGluR1 has a critical role in long-term synaptic depression (LTD) at parallel fiber (PF)-PC synapses, and in developmental elimination of climbing fiber (CF)-PC synapses. mGluR1a, a predominant splice variant in PCs, has a long carboxyl (C)-terminal domain that interacts with Homer scaffolding proteins. Cerebellar roles of the C-terminal domain at both synaptic and behavior levels remain poorly understood. To address this question, we introduced a short variant, mGluR1b, which lacks this domain into PCs of mGluR1-knock-out (KO) mice (mGluR1b-rescue mice). In mGluR1b-rescue mice, mGluR1b showed dispersed perisynaptic distribution in PC spines. Importantly, mGluR1b-rescue mice exhibited impairments in inositol 1,4,5-trisphosphate receptor (IP3R)-mediated Ca(2+) release, CF synapse elimination, LTD induction, and delay eyeblink conditioning: they showed normal transient receptor potential canonical (TRPC) currents and normal motor coordination. In contrast, PC-specific rescue of mGluR1a restored all cerebellar defects of mGluR1-KO mice. We conclude that the long C-terminal domain of mGluR1a is required for the proper perisynaptic targeting of mGluR1, IP3R-mediated Ca(2+) release, CF synapse elimination, LTD, and motor learning, but not for TRPC currents and motor coordination.
Collapse
|
222
|
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
|
223
|
Yao H, Bethel-Brown C, Niu F, Yang L, Peng F, Buch S. Yin and Yang of PDGF-mediated signaling pathway in the context of HIV infection and drug abuse. J Neuroimmune Pharmacol 2014; 9:161-7. [PMID: 23784143 PMCID: PMC3865168 DOI: 10.1007/s11481-013-9481-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 05/28/2013] [Indexed: 01/06/2023]
Abstract
The control and eradication of neurological complications associated with AIDS continues to be an important goal in efforts toward improving the well being of HIV-infected patients. Although combined antiretroviral therapies have contributed significantly to increasing the longevity of patients by suppressing the virus burden in the systemic compartments, the prevalence of HIV-associated neurological disorders continues to be on the rise. This in turn, leads to an impaired quality of life of the infected individuals who continue to suffer from mild to moderate cognitive decline and memory loss. Developing therapeutic interventions that reverse neuronal injury in the context of HIV infection, is thus of paramount importance in the field. Our previous studies have demonstrated that platelet-derived growth factor (PDGF) has a neuroprotective potential against HIV envelope protein gp120 and Tat. Paradoxically, PDGF is also a cerebrovascular permeant with deleterious effects on the blood-brain barrier resulting in increased influx of monocytes in the CNS. Herein, we review the opposing roles of PDGF in the context of HIV-associated neurodegenerative disorder (HAND).
Collapse
Affiliation(s)
- Honghong Yao
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198
| | - Crystal Bethel-Brown
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198
| | - Fang Niu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198
| | - Lu Yang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198
| | - Fuwang Peng
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
224
|
Distinct and simultaneously active plasticity mechanisms in mouse hippocampus during different phases of Morris water maze training. Brain Struct Funct 2014; 220:1273-90. [PMID: 24562414 DOI: 10.1007/s00429-014-0722-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 01/28/2014] [Indexed: 01/30/2023]
Abstract
Although the Morris water maze (MWM) is the most frequently used protocol to examine hippocampus-dependent learning in mice, not much is known about the spatio-temporal dynamics of underlying plasticity processes. Here, we studied molecular and cellular hippocampal plasticity mechanisms during early and late phases of spatial learning in the MWM. Quantitative in situ hybridization for the immediate early genes zif268 and Homer1a (H1a) revealed phase-dependent differences in their expression between areas CA1 and CA3. During the initial learning phase, CA1 expression levels of the molecular plasticity marker H1a, but not of the activity reporter gene zif268, were related to task proficiency; whereas no learning-specific changes could be detected in CA3. Simultaneously, the ratio of surface-expressed NMDAR subunits NR2A and NR2B was downregulated as measured by acute slice biotinylation assay, while the total number of surface NMDARs was unaltered. When intrinsic 'somatic' and synaptic plasticity in the CA1-region of hippocampal slices were examined, we found that early learning promotes intrinsic neuronal plasticity as manifested by a reduction of spike frequency adaptation and postburst afterhyperpolarization. At the synaptic level, however, maintenance of long-term potentiation (LTP) in all learning groups was impaired which is most likely due to 'intrinsic' learning-induced LTP which occluded any further electrically induced LTP. Late learning, in contrast, was characterized by re-normalized H1a, NR2A and NR2B expression and neuronal firing, yet a further strengthening of learning-induced LTP. Together, our data support a precisely timed cascade of complex molecular and subcellular transformations occurring from early to late MWM learning.
Collapse
|
225
|
D'Antoni S, Spatuzza M, Bonaccorso CM, Musumeci SA, Ciranna L, Nicoletti F, Huber KM, Catania MV. Dysregulation of group-I metabotropic glutamate (mGlu) receptor mediated signalling in disorders associated with Intellectual Disability and Autism. Neurosci Biobehav Rev 2014; 46 Pt 2:228-41. [PMID: 24548786 DOI: 10.1016/j.neubiorev.2014.02.003] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 01/13/2014] [Accepted: 02/06/2014] [Indexed: 12/11/2022]
Abstract
Activation of group-I metabotropic glutamate receptors, mGlu1 and mGlu5, triggers a variety of signalling pathways in neurons and glial cells, which are differently implicated in synaptic plasticity. The earliest and much of key studies discovered abnormal mGlu5 receptor function in Fragile X syndrome (FXS) mouse models which then motivated more recent work that finds mGlu5 receptor dysfunction in related disorders such as intellectual disability (ID), obsessive-compulsive disorder (OCD) and autism. Therefore, mGlu1/5 receptor dysfunction may represent a common aetiology of these complex diseases. Furthermore, many studies have focused on dysregulation of mGlu5 signalling to synaptic protein synthesis. However, emerging evidence finds abnormal mGlu5 receptor interactions with its scaffolding proteins in FXS which results in mGlu5 receptor dysfunction and phenotypes independent of signalling to protein synthesis. Finally, both an increased and reduced mGlu5 functioning seem to be associated with ID and autism spectrum disorders, with important consequences for potential treatment of these developmental disorders.
Collapse
Affiliation(s)
- Simona D'Antoni
- Institute of Neurological Sciences, the National Research Council of Italy (CNR), Catania, Italy
| | - Michela Spatuzza
- Institute of Neurological Sciences, the National Research Council of Italy (CNR), Catania, Italy
| | | | | | - Lucia Ciranna
- Department of Biomedical Sciences, section of Physiology, University of Catania, Italy
| | - Ferdinando Nicoletti
- IRCCS Neuromed, Pozzilli (IS), Italy; University of Rome La Sapienza, Rome, Italy
| | - Kimberly M Huber
- University of Texas Southwestern Medical Center, Department of Neuroscience, Dallas, TX 75390-9111, USA
| | - Maria Vincenza Catania
- Institute of Neurological Sciences, the National Research Council of Italy (CNR), Catania, Italy; IRCCS Oasi Maria SS, Troina (EN), Italy.
| |
Collapse
|
226
|
Schneider A, Rogalewski A, Wafzig O, Kirsch F, Gretz N, Krüger C, Diederich K, Pitzer C, Laage R, Plaas C, Vogt G, Minnerup J, Schäbitz WR. Forced arm use is superior to voluntary training for motor recovery and brain plasticity after cortical ischemia in rats. EXPERIMENTAL & TRANSLATIONAL STROKE MEDICINE 2014; 6:3. [PMID: 24528872 PMCID: PMC3937028 DOI: 10.1186/2040-7378-6-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 01/26/2014] [Indexed: 12/24/2022]
Abstract
Background and purpose Both the immobilization of the unaffected arm combined with physical therapy (forced arm use, FAU) and voluntary exercise (VE) as model for enriched environment are promising approaches to enhance recovery after stroke. The genomic mechanisms involved in long-term plasticity changes after different means of rehabilitative training post-stroke are largely unexplored. The present investigation explored the effects of these physical therapies on behavioral recovery and molecular markers of regeneration after experimental ischemia. Methods 42 Wistar rats were randomly treated with either forced arm use (FAU, 1-sleeve plaster cast onto unaffected limb at 8/10 days), voluntary exercise (VE, connection of a freely accessible running wheel to cage), or controls with no access to a running wheel for 10 days starting at 48 hours after photothrombotic stroke of the sensorimotor cortex. Functional outcome was measured using sensorimotor test before ischemia, after ischemia, after the training period of 10 days, at 3 and 4 weeks after ischemia. Global gene expression changes were assessed from the ipsi- and contralateral cortex and the hippocampus. Results FAU-treated animals demonstrated significantly improved functional recovery compared to the VE-treated group. Both were superior to cage control. A large number of genes are altered by both training paradigms in the ipsi- and contralateral cortex and the hippocampus. Overall, the extent of changes observed correlated well with the functional recovery obtained. One category of genes overrepresented in the gene set is linked to neuronal plasticity processes, containing marker genes such as the NMDA 2a receptor, PKC ζ, NTRK2, or MAP 1b. Conclusions We show that physical training after photothrombotic stroke significantly and permanently improves functional recovery after stroke, and that forced arm training is clearly superior to voluntary running training. The behavioral outcomes seen correlate with patterns and extent of gene expression changes in all brain areas examined. We propose that physical training induces a fundamental change in plasticity-relevant gene expression in several brain regions that enables recovery processes. These results contribute to the debate on optimal rehabilitation strategies, and provide a valuable source of molecular entry points for future pharmacological enhancement of recovery.
Collapse
Affiliation(s)
| | - Andreas Rogalewski
- Neurology Department Bethel EVKB Bielefeld and Dept. of Neurology, University of Muenster, Muenster, Germany
| | | | | | - Norbert Gretz
- Ctr. for Med. Res., Fac. for Clin. Medicine, University of Heidelberg, Mannheim, Germany
| | | | - Kai Diederich
- Neurology Department Bethel EVKB Bielefeld and Dept. of Neurology, University of Muenster, Muenster, Germany
| | | | | | | | | | - Jens Minnerup
- Neurology Department Bethel EVKB Bielefeld and Dept. of Neurology, University of Muenster, Muenster, Germany
| | - Wolf-Rüdiger Schäbitz
- Neurology Department Bethel EVKB Bielefeld and Dept. of Neurology, University of Muenster, Muenster, Germany
| |
Collapse
|
227
|
Wagner KV, Häusl AS, Pöhlmann ML, Hartmann J, Labermaier C, Müller MB, Schmidt MV. Hippocampal Homer1 levels influence motivational behavior in an operant conditioning task. PLoS One 2014; 9:e85975. [PMID: 24465821 PMCID: PMC3897610 DOI: 10.1371/journal.pone.0085975] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 12/03/2013] [Indexed: 11/23/2022] Open
Abstract
Loss of motivation and learning impairments are commonly accepted core symptoms of psychiatric disorders such as depression and schizophrenia. Reward-motivated learning is dependent on the hippocampal formation but the molecular mechanisms that lead to functional incentive motivation in this brain region are still largely unknown. Recent evidence implicates neurotransmission via metabotropic glutamate receptors and Homer1, their interaction partner in the postsynaptic density, in drug addiction and motivational learning. As previous reports mainly focused on the prefrontal cortex and the nucleus accumbens, we now investigated the role of hippocampal Homer1 in operant reward learning in the present study. We therefore tested either Homer1 knockout mice or mice that overexpress Homer1 in the hippocampus in an operant conditioning paradigm. Our results show that deletion of Homer1 leads to a diverging phenotype that either displays an inability to perform the task or outstanding hyperactivity in both learning and motivational sessions. Due to the apparent bimodal distribution of this phenotype, the overall effect of Homer1 deletion in this paradigm is not significantly altered. Overexpression of hippocampal Homer1 did not lead to a significantly altered learning performance in any stage of the testing paradigm, yet may subtly contribute to emerging motivational deficits. Our results indicate an involvement of Homer1-mediated signaling in the hippocampus in motivation-based learning tasks and encourage further investigations regarding the specific molecular underpinnings of the phenotypes observed in this study. We also suggest to cautiously interpret the results of this and other studies regarding the phenotype following Homer1 manipulations in animals, since their behavioral phenotype appears to be highly diverse. Future studies would benefit from larger group sizes that would allow splitting the experimental groups in responders and non-responders.
Collapse
Affiliation(s)
- Klaus V. Wagner
- Research Group Neurobiology of Stress, Max Planck Institute of Psychiatry, Munich, Bavaria, Germany
- * E-mail:
| | - Alexander S. Häusl
- Research Group Neurobiology of Stress, Max Planck Institute of Psychiatry, Munich, Bavaria, Germany
| | - Max L. Pöhlmann
- Research Group Neurobiology of Stress, Max Planck Institute of Psychiatry, Munich, Bavaria, Germany
| | - Jakob Hartmann
- Research Group Neurobiology of Stress, Max Planck Institute of Psychiatry, Munich, Bavaria, Germany
| | - Christiana Labermaier
- Research Group Neurobiology of Stress, Max Planck Institute of Psychiatry, Munich, Bavaria, Germany
| | - Marianne B. Müller
- Research Group Neurobiology of Stress, Max Planck Institute of Psychiatry, Munich, Bavaria, Germany
| | - Mathias V. Schmidt
- Research Group Neurobiology of Stress, Max Planck Institute of Psychiatry, Munich, Bavaria, Germany
| |
Collapse
|
228
|
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
|
229
|
Fuxe K, Borroto-Escuela DO, Ciruela F, Guidolin D, Agnati LF. Receptor-receptor interactions in heteroreceptor complexes: a new principle in biology. Focus on their role in learning and memory. ACTA ACUST UNITED AC 2014. [DOI: 10.7243/2052-6946-2-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
230
|
Moore ED, Kooshki M, Wheeler KT, Metheny-Barlow LJ, Robbins ME. Differential expression of Homer1a in the hippocampus and cortex likely plays a role in radiation-induced brain injury. Radiat Res 2013; 181:21-32. [PMID: 24377717 DOI: 10.1667/rr13475.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Fractionated partial or whole-brain irradiation is the primary treatment for metastatic brain tumors. Despite reducing tumor burden and increasing lifespan, progressive, irreversible cognitive impairment occurs in >50% of the patients who survive >6 months after fractionated whole-brain irradiation. The exact mechanism(s) responsible for this radiation-induced brain injury are unknown; however, preclinical studies suggest that radiation modulates the extracellular receptor kinase signaling pathway, which is associated with cognitive impairment in many neurological diseases. In the study reported here, we demonstrated that the extracellular receptor kinase transcriptionally-regulated early response gene, Homer1a, was up-regulated transiently in the hippocampus and down-regulated in the cortex of young adult male Fischer 344 X Brown Norway rats at 48 h after 40 Gy of fractionated whole-brain irradiation. Two months after fractionated whole-brain irradiation, these changes in Homer1a expression correlated with a down-regulation of the hippocampal glutamate receptor 1 and protein kinase Cγ, and an up-regulation of cortical glutamate receptor 1 and protein kinase Cγ. Two drugs that prevent radiation-induced cognitive impairment in rats, the angiotensin type-1 receptor blocker, L-158,809, and the angiotensin converting enzyme inhibitor, ramipril, reversed the fractionated whole-brain irradiation-induced Homer1a expression at 48 h in the hippocampus and cortex and restored glutamate receptor 1 and protein kinase Cγ to the levels in sham-irradiated controls at 2 months after fractionated whole-brain irradiation. These data indicate that Homer1a is, (1) a brain region specific regulator of radiation-induced brain injury, including cognitive impairment and (2) potentially a druggable target for preventing it.
Collapse
|
231
|
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
|
232
|
Lee H, Lee EJ, Song YS, Kim E. Long-term depression-inducing stimuli promote cleavage of the synaptic adhesion molecule NGL-3 through NMDA receptors, matrix metalloproteinases and presenilin/γ-secretase. Philos Trans R Soc Lond B Biol Sci 2013; 369:20130158. [PMID: 24298159 PMCID: PMC3843889 DOI: 10.1098/rstb.2013.0158] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Long-term depression (LTD) reduces the functional strength of excitatory synapses through mechanisms that include the removal of AMPA glutamate receptors from the postsynaptic membrane. LTD induction is also known to result in structural changes at excitatory synapses, including the shrinkage of dendritic spines. Synaptic adhesion molecules are thought to contribute to the development, function and plasticity of neuronal synapses largely through their trans-synaptic adhesions. However, little is known about how synaptic adhesion molecules are altered during LTD. We report here that NGL-3 (netrin-G ligand-3), a postsynaptic adhesion molecule that trans-synaptically interacts with the LAR family of receptor tyrosine phosphatases and intracellularly with the postsynaptic scaffolding protein PSD-95, undergoes a proteolytic cleavage process. NGL-3 cleavage is induced by NMDA treatment in cultured neurons and low-frequency stimulation in brain slices and requires the activities of NMDA glutamate receptors, matrix metalloproteinases (MMPs) and presenilin/γ-secretase. These results suggest that NGL-3 is a novel substrate of MMPs and γ-secretase and that NGL-3 cleavage may regulate synaptic adhesion during LTD.
Collapse
Affiliation(s)
- Hyejin Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), , Daejeon 305-701, Korea
| | | | | | | |
Collapse
|
233
|
Abstract
Among the largest cells in the body, neurons possess an immense surface area and intricate geometry that poses many unique cell biological challenges. This morphological complexity is critical for neural circuit formation and enables neurons to compartmentalize cell-cell communication and local intracellular signalling to a degree that surpasses other cell types. The adaptive plastic properties of neurons, synapses and circuits have been classically studied by measurement of electrophysiological properties, ionic conductances and excitability. Over the last 15 years, the field of synaptic and neural electrophysiology has collided with neuronal cell biology to produce a more integrated understanding of how these remarkable highly differentiated cells utilize common eukaryotic cellular machinery to decode, integrate and propagate signals in the nervous system. The present article gives a very brief and personal overview of the organelles and trafficking machinery of neuronal dendrites and their role in dendritic and synaptic plasticity.
Collapse
Affiliation(s)
- Michael D Ehlers
- *Neuroscience Research Unit, Pfizer Worldwide Research and Development, 700 Main Street, Cambridge, MA 02139, U.S.A
| |
Collapse
|
234
|
Park JM, Hu JH, Milshteyn A, Zhang PW, Moore CG, Park S, Datko MC, Domingo RD, Reyes CM, Wang XJ, Etzkorn FA, Xiao B, Szumlinski KK, Kern D, Linden DJ, Worley PF. A prolyl-isomerase mediates dopamine-dependent plasticity and cocaine motor sensitization. Cell 2013; 154:637-50. [PMID: 23911326 DOI: 10.1016/j.cell.2013.07.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 04/14/2013] [Accepted: 07/01/2013] [Indexed: 01/02/2023]
Abstract
Synaptic plasticity induced by cocaine and other drugs underlies addiction. Here we elucidate molecular events at synapses that cause this plasticity and the resulting behavioral response to cocaine in mice. In response to D1-dopamine-receptor signaling that is induced by drug administration, the glutamate-receptor protein metabotropic glutamate receptor 5 (mGluR5) is phosphorylated by microtubule-associated protein kinase (MAPK), which we show potentiates Pin1-mediated prolyl-isomerization of mGluR5 in instances where the product of an activity-dependent gene, Homer1a, is present to enable Pin1-mGluR5 interaction. These biochemical events potentiate N-methyl-D-aspartate receptor (NMDAR)-mediated currents that underlie synaptic plasticity and cocaine-evoked motor sensitization as tested in mice with relevant mutations. The findings elucidate how a coincidence of signals from the nucleus and the synapse can render mGluR5 accessible to activation with consequences for drug-induced dopamine responses and point to depotentiation at corticostriatal synapses as a possible therapeutic target for treating addiction.
Collapse
Affiliation(s)
- Joo Min Park
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
235
|
Li Z, Qiu HY, Jiao Y, Cen JN, Fu CM, Hu SY, Zhu MQ, Wu DP, Qi XF. Growth and differentiation effects of Homer3 on a leukemia cell line. Asian Pac J Cancer Prev 2013; 14:2525-8. [PMID: 23725168 DOI: 10.7314/apjcp.2013.14.4.2525] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The Homer protein family, also known as the family of cytoplasmic scaffolding proteins, which include three subtypes (Homer1, Homer2, Homer3). Homer3 can regulate transcription and play a very important role in the differentiation and development for some tissues (e.g. muscle and nervous systems). The current studies showed that Homer3 abnormal expression changes in acute myeloid leukemia (AML). Forced expression of Homer3 in transfected K562 cells inhibited proliferation, influenced the cell cycle profile, affected apoptosis induced by As2O3 through inhibition of Bcl2 expression, and also promoted cell differentiation induced by 12-O-tetra decanoylphorbol-acetate (TPA). These results showed that Homer3 is a novel gene which plays a certain role in the occurrence and development of AML.
Collapse
Affiliation(s)
- Zheng Li
- Leukemia Research Division, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of the Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
236
|
Hermey G, Mahlke C, Gutzmann JJ, Schreiber J, Blüthgen N, Kuhl D. Genome-wide profiling of the activity-dependent hippocampal transcriptome. PLoS One 2013; 8:e76903. [PMID: 24146943 PMCID: PMC3798291 DOI: 10.1371/journal.pone.0076903] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 08/25/2013] [Indexed: 01/21/2023] Open
Abstract
Activity-dependent gene expression is central for sculpting neuronal connectivity in the brain. Despite the importance for synaptic plasticity, a comprehensive analysis of the temporal changes in the transcriptomic response to neuronal activity is lacking. In a genome wide survey we identified genes that were induced at 1, 4, 8, or 24 hours following neuronal activity in the hippocampus. According to their distinct expression kinetics we assigned these genes to five clusters, each containing approximately 200 genes. Using in situ hybridizations the regulated expression of 24 genes was validated. Apart from known activity-dependent genes our study reveals a large number of unknown induced genes with distinct expression kinetics. Among these we identified several genes with complex temporal expression patterns. Furthermore, our study provides examples for activity-induced exon switching in the coding region of genes and activity-induced alternative splicing of the 3'-UTR. One example is Zwint. In contrast to the constitutively expressed variant, the induced Zwint transcript harbors multiple regulatory elements in the 3'-UTR. Taken together, our study provides a comprehensive analysis of the transcriptomic response to neuronal activity and sheds new light on expression kinetics and alternative splicing events.
Collapse
Affiliation(s)
- Guido Hermey
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- * E-mail: (GH); (DK)
| | - Claudia Mahlke
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jakob J. Gutzmann
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jörg Schreiber
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Nils Blüthgen
- Institute for Theoretical Biology and Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dietmar Kuhl
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- * E-mail: (GH); (DK)
| |
Collapse
|
237
|
Kaja S, Sumien N, Borden PK, Khullar N, Iqbal M, Collins JL, Forster MJ, Koulen P. Homer-1a immediate early gene expression correlates with better cognitive performance in aging. AGE (DORDRECHT, NETHERLANDS) 2013; 35:1799-1808. [PMID: 23054826 PMCID: PMC3776093 DOI: 10.1007/s11357-012-9479-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 09/13/2012] [Indexed: 05/31/2023]
Abstract
The molecular mechanisms underlying cognitive decline during healthy aging remain largely unknown. Utilizing aged wild-type C57BL/6 mice as a model for normal aging, we tested the hypothesis that cognitive performance, memory, and learning as assessed in established behavioral testing paradigms are correlated with the differential expression of isoforms of the Homer family of synaptic scaffolding proteins. Here we describe a loss of cognitive and motor function that occurs when Homer-1a/Vesl-1S protein levels drop during aging. Our data describe a novel mechanism of age-related synaptic changes contributing to loss of biological function, spatial learning, and memory formation as well as motor coordination, with the dominant negative uncoupler of synaptic protein clustering, Homer-1a/Vesl-1S, as a potential target for the prophylaxis and treatment of age-related cognitive decline.
Collapse
Affiliation(s)
- Simon Kaja
- />Department Ophthalmology and Vision Research Center, School of Medicine, University of Missouri—Kansas City, 2411 Holmes St., Kansas City, MO 64108 USA
| | - Nathalie Sumien
- />Department of Pharmacology and Neuroscience and Institute for Aging and Alzheimer’s Disease Research, University of North Texas Health Science Center, Fort Worth, TX 76107 USA
| | - Priscilla K. Borden
- />Department Ophthalmology and Vision Research Center, School of Medicine, University of Missouri—Kansas City, 2411 Holmes St., Kansas City, MO 64108 USA
| | - Nitasha Khullar
- />Department Ophthalmology and Vision Research Center, School of Medicine, University of Missouri—Kansas City, 2411 Holmes St., Kansas City, MO 64108 USA
| | - Maaz Iqbal
- />Department Ophthalmology and Vision Research Center, School of Medicine, University of Missouri—Kansas City, 2411 Holmes St., Kansas City, MO 64108 USA
| | - Julie L. Collins
- />Department Ophthalmology and Vision Research Center, School of Medicine, University of Missouri—Kansas City, 2411 Holmes St., Kansas City, MO 64108 USA
| | - Michael J. Forster
- />Department of Pharmacology and Neuroscience and Institute for Aging and Alzheimer’s Disease Research, University of North Texas Health Science Center, Fort Worth, TX 76107 USA
| | - Peter Koulen
- />Department Ophthalmology and Vision Research Center, School of Medicine, University of Missouri—Kansas City, 2411 Holmes St., Kansas City, MO 64108 USA
- />Department of Basic Medical Science, School of Medicine, University of Missouri—Kansas City, 2411 Holmes St., Kansas City, MO 64108 USA
| |
Collapse
|
238
|
Chen T, Yang YF, Luo P, Liu W, Dai SH, Zheng XR, Fei Z, Jiang XF. Homer1 knockdown protects dopamine neurons through regulating calcium homeostasis in an in vitro model of Parkinson's disease. Cell Signal 2013; 25:2863-70. [PMID: 24036210 DOI: 10.1016/j.cellsig.2013.09.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 08/15/2013] [Accepted: 09/02/2013] [Indexed: 11/26/2022]
Abstract
Homer1 protein is an important scaffold protein at postsynaptic density and has been demonstrated to play a central role in calcium signaling in the central nervous system. The aim of this study was to investigate the effects of Homer1 knockdown on MPP(+) induced neuronal injury in cultured dopamine (DA) neurons. We found that down-regulating Homer1 expression with specific small interfering RNA (siRNA) significantly suppressed LDH release, reduced Propidium iodide (PI) or Hoechst staining, increased the number of tyrosine hydroxylase (TH) positive cells and DA uptake, and attenuated apoptotic and necrotic cell death after MPP(+) injury. Homer1 knockdown decreased intracellular reactive oxygen species (ROS) generation through inhibition of intracellular calcium overload, but did not affect the endogenous antioxidant enzyme activities. Calcium imaging was used to examine the changes of intracellular Ca(2+) concentration ([Ca(2+)]cyt) and Ca(2+) in endoplasmic reticulum (ER) ([Ca(2+)]ER), and the results showed that Homer1 siRNA transfection attenuated ER Ca(2+) release up to 120min after MPP(+) injury. Furthermore, decrease of [Ca(2+)]cyt induced by Homer1 knockdown in MPP(+) treated neurons was further enhanced by NMDA receptor antagonists MK-801 and AP-5, but not canonical transient receptor potential (TRPC) channel antagonist SKF-96365. l-type calcium antagonist isradipine but not nimodipine further inhibited intracellular calcium overload after MPP(+) insult in Homer1 down-regulated neurons. These results suggest that Homer1 knockdown has protective effects against neuronal injury in in vitro PD model by reducing calcium overload mediated ROS generation, and this protection may be dependent at least in part on the regulatory effects on the function of calcium channels in both plasma membrane and ER.
Collapse
Affiliation(s)
- Tao Chen
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Fourth, Military Medical University, Xi'an, Shaanxi 710032, China; Department of Neurosurgery, The 123th Hospital of PLA, Bengbu, Anhui 233000, China
| | | | | | | | | | | | | | | |
Collapse
|
239
|
Willard SS, Koochekpour S. Glutamate signaling in benign and malignant disorders: current status, future perspectives, and therapeutic implications. Int J Biol Sci 2013; 9:728-42. [PMID: 23983606 PMCID: PMC3753409 DOI: 10.7150/ijbs.6475] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 06/15/2013] [Indexed: 11/05/2022] Open
Abstract
Glutamate, a nonessential amino acid, is the major excitatory neurotransmitter in the central nervous system. As such, glutamate has been shown to play a role in not only neural processes, such as learning and memory, but also in bioenergetics, biosynthetic and metabolic oncogenic pathways. Glutamate has been the target of intense investigation for its involvement not only in the pathogenesis of benign neurodegenerative diseases (NDDs) such as Parkinson's disease, Alzheimer's disease, schizophrenia, multiple sclerosis, and amyotropic lateral sclerosis (ALS), but also in carcinogenesis and progression of malignant diseases. In addition to its intracellular activities, glutamate in secreted form is a phylogenetically conserved cell signaling molecule. Glutamate binding activates multiple major receptor families including the metabotropic glutamate receptors (mGluRs) and ionotropic glutamate receptors (iGluRs), both of which have been implicated in various signaling pathways in cancer. Inhibition of extracellular glutamate release or glutamate receptor activation via competitive or non-competitive antagonists decreases growth, migration and invasion and induces apoptosis in breast cancer, melanoma, glioma and prostate cancer cells. In this review, we discuss the current state of glutamate signaling research as it relates to benign and malignant diseases. In addition, we provide a synopsis of clinical trials using glutamate antagonists for the treatment of NDD and malignant diseases. We conclude that in addition to its potential role as a metabolic biomarker, glutamate receptors and glutamate-initiated signaling pathways may provide novel therapeutic opportunities for cancer.
Collapse
Affiliation(s)
- Stacey S Willard
- Departments of Cancer Genetics and Urology, Center for Genetics and Pharmacology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | | |
Collapse
|
240
|
Salanova M, Volpe P, Blottner D. Homer protein family regulation in skeletal muscle and neuromuscular adaptation. IUBMB Life 2013; 65:769-76. [DOI: 10.1002/iub.1198] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 06/03/2013] [Accepted: 06/24/2013] [Indexed: 01/28/2023]
Affiliation(s)
- Michele Salanova
- Department of Vegetative Anatomy and Center of Space Medicine Berlin (ZWMB); Neuromuscular Group; Charité Universitätsmedizin Berlin; Berlin; Germany
| | - Pompeo Volpe
- Dipartimento di Scienze Biomediche; Università di Padova, Italy; C.N.R. Institute of Neuroscience; Padova; Italy
| | - Dieter Blottner
- Department of Vegetative Anatomy and Center of Space Medicine Berlin (ZWMB); Neuromuscular Group; Charité Universitätsmedizin Berlin; Berlin; Germany
| |
Collapse
|
241
|
Martínez-Rivera A, Rodríguez-Borrero E, Matías-Alemán M, Montalvo-Acevedo A, Guerrero-Figuereo K, Febo-Rodríguez LJ, Morales-Rivera A, Maldonado-Vlaar CS. Metabotropic glutamate receptor 5 within nucleus accumbens shell modulates environment-elicited cocaine conditioning expression. Pharmacol Biochem Behav 2013; 110:154-60. [PMID: 23850523 DOI: 10.1016/j.pbb.2013.06.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 06/05/2013] [Accepted: 06/23/2013] [Indexed: 10/26/2022]
Abstract
The metabotropic glutamate receptors 5 (mGluRs5) within the Nucleus Accumbens (NAc) have been implicated in the modulation of psychostimulant reward. We hypothesized that blockade of mGluR5 within the NAc shell would impair cocaine conditioning in rats. For this study, animals were implanted with cannulae within the NAc shell, and separate groups were exposed to a multimodal environment within activity chambers that signaled cocaine (cocaine-paired) or saline (controls, cocaine-unpaired) injections. Prior to placing the animals in the chambers, rats received systemic intraperitoneal injections of saline or cocaine for 10 consecutive sessions. In the test session (D12), animals were exposed to the multimodal environment without any cocaine or saline pre-treatment. Before placing the rats in the chambers, separate groups of animals were infused within the NAc shell with 2.5, 12 or 25 nmol/0.5 μl/side of 2-methyl-6-(phenylethynyl) pyridine (MPEP), an antagonist of mGluR5 or with vehicle. Blockade of the mGluR5 subtype at a 2.5 nmol dose showed no significant difference in either the ambulatory distance (AD) or the vertical plane move time (VPT). In contrast, mGluR5 blockade at 12 nmol and 25 nmol decreased conditioned locomotion in the cocaine-paired groups. An association of the environmental cues with the effects of cocaine implies the involvement of memory process during the conditioning response. Our results suggest that mGluR5 within the NAc shell could be modulating the expression of memory related to the association of environmental cues with the effects of cocaine. We suggest that mGluR5 could be taking into account to further studies related with cocaine exposure and cocaine addiction treatments.
Collapse
|
242
|
Postnatal development ofHomer1ain the rat hippocampus. Hippocampus 2013; 23:890-902. [DOI: 10.1002/hipo.22146] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2013] [Indexed: 01/11/2023]
|
243
|
Pribiag H, Stellwagen D. Neuroimmune regulation of homeostatic synaptic plasticity. Neuropharmacology 2013; 78:13-22. [PMID: 23774138 DOI: 10.1016/j.neuropharm.2013.06.008] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 05/28/2013] [Accepted: 06/02/2013] [Indexed: 01/08/2023]
Abstract
Homeostatic synaptic plasticity refers to a set of negative-feedback mechanisms that are used by neurons to maintain activity within a functional range. While it is becoming increasingly clear that homeostatic regulation of synapse function is a key principle in the nervous system, the molecular details of this regulation are only beginning to be uncovered. Recent evidence implicates molecules classically associated with the peripheral immune system in the modulation of homeostatic synaptic plasticity. In particular, the pro-inflammatory cytokine TNFα, class I major histocompatibility complex, and neuronal pentraxin 2 are essential in the regulation of the compensatory synaptic response that occurs in response to prolonged neuronal inactivity. This review will present and discuss current evidence implicating neuroimmune molecules in the homeostatic regulation of synapse function. This article is part of the Special Issue entitled 'Homeostatic Synaptic Plasticity'.
Collapse
Affiliation(s)
- Horia Pribiag
- Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montreal General Hospital, L7-132, 1650 Cedar Av, Montreal, QC H3G 1A4, Canada
| | - David Stellwagen
- Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montreal General Hospital, L7-132, 1650 Cedar Av, Montreal, QC H3G 1A4, Canada.
| |
Collapse
|
244
|
Oku S, Takahashi N, Fukata Y, Fukata M. In silico screening for palmitoyl substrates reveals a role for DHHC1/3/10 (zDHHC1/3/11)-mediated neurochondrin palmitoylation in its targeting to Rab5-positive endosomes. J Biol Chem 2013; 288:19816-29. [PMID: 23687301 DOI: 10.1074/jbc.m112.431676] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Protein palmitoylation, a common post-translational lipid modification, plays an important role in protein trafficking and functions. Recently developed palmitoyl-proteomic methods identified many novel substrates. However, the whole picture of palmitoyl substrates has not been clarified. Here, we performed global in silico screening using the CSS-Palm 2.0 program, free software for prediction of palmitoylation sites, and selected 17 candidates as novel palmitoyl substrates. Of the 17 candidates, 10 proteins, including 6 synaptic proteins (Syd-1, transmembrane AMPA receptor regulatory protein (TARP) γ-2, TARP γ-8, cornichon-2, Ca(2+)/calmodulin-dependent protein kinase IIα, and neurochondrin (Ncdn)/norbin), one focal adhesion protein (zyxin), two ion channels (TRPM8 and TRPC1), and one G-protein-coupled receptor (orexin 2 receptor), were palmitoylated. Using the DHHC palmitoylating enzyme library, we found that all tested substrates were palmitoylated by the Golgi-localized DHHC3/7 subfamily. Ncdn, a regulator for neurite outgrowth and synaptic plasticity, was robustly palmitoylated by the DHHC1/10 (zDHHC1/11; z1/11) subfamily, whose substrate has not yet been reported. As predicted by CSS-Palm 2.0, Cys-3 and Cys-4 are the palmitoylation sites for Ncdn. Ncdn was specifically localized in somato-dendritic regions, not in the axon of rat cultured neurons. Stimulated emission depletion microscopy revealed that Ncdn was localized to Rab5-positive early endosomes in a palmitoylation-dependent manner, where DHHC1/10 (z1/11) were also distributed. Knockdown of DHHC1, -3, or -10 (z11) resulted in the loss of Ncdn from Rab5-positive endosomes. Thus, through in silico screening, we demonstrate that Ncdn and the DHHC1/10 (z1/11) and DHHC3/7 subfamilies are novel palmitoyl substrate-enzyme pairs and that Ncdn palmitoylation plays an essential role in its specific endosomal targeting.
Collapse
Affiliation(s)
- Shinichiro Oku
- Division of Membrane Physiology, Department of Cell Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | | | | | | |
Collapse
|
245
|
Negative feedback regulation of Homer 1a on norepinephrine-dependent cardiac hypertrophy. Exp Cell Res 2013; 319:1804-1814. [PMID: 23664835 DOI: 10.1016/j.yexcr.2013.04.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Revised: 04/22/2013] [Accepted: 04/28/2013] [Indexed: 01/14/2023]
Abstract
Homers are scaffolding proteins that modulate diverse cell functions being able to assemble signalling complexes. In this study, the presence, sub-cellular distribution and function of Homer 1 was investigated. Homer 1a and Homer 1b/c are constitutively expressed in cardiac muscle of both mouse and rat and in HL-1 cells, a cardiac cell line. As judged by confocal immunofluorescence microscopy, Homer 1a displays sarcomeric and peri-nuclear localization. In cardiomyocytes and cultured HL-1 cells, the hypertrophic agonist norepinephrine (NE) induces α1-adrenergic specific Homer 1a over-expression, with a two-to-three-fold increase within 1h, and no up-regulation of Homer 1b/c, as judged by Western blot and qPCR. In HL-1 cells, plasmid-driven over-expression of Homer 1a partially antagonizes activation of ERK phosphorylation and ANF up-regulation, two well-established, early markers of hypertrophy. At the morphometric level, NE-induced increase of cell size is likewise and partially counteracted by exogenous Homer 1a. Under the same experimental conditions, Homer 1b/c does not have any effect on ANF up-regulation nor on cell hypertrophy. Thus, Homer 1a up-regulation is associated to early stages of cardiac hypertrophy and appears to play a negative feedback regulation on molecular transducers of hypertrophy.
Collapse
|
246
|
Bhattacharjee A, Oeemig JS, Kolodziejczyk R, Meri T, Kajander T, Lehtinen MJ, Iwaï H, Jokiranta TS, Goldman A. Structural basis for complement evasion by Lyme disease pathogen Borrelia burgdorferi. J Biol Chem 2013; 288:18685-95. [PMID: 23658013 DOI: 10.1074/jbc.m113.459040] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Borrelia burgdorferi spirochetes that cause Lyme borreliosis survive for a long time in human serum because they successfully evade the complement system, an important arm of innate immunity. The outer surface protein E (OspE) of B. burgdorferi is needed for this because it recruits complement regulator factor H (FH) onto the bacterial surface to evade complement-mediated cell lysis. To understand this process at the molecular level, we used a structural approach. First, we solved the solution structure of OspE by NMR, revealing a fold that has not been seen before in proteins involved in complement regulation. Next, we solved the x-ray structure of the complex between OspE and the FH C-terminal domains 19 and 20 (FH19-20) at 2.83 Å resolution. The structure shows that OspE binds FH19-20 in a way similar to, but not identical with, that used by endothelial cells to bind FH via glycosaminoglycans. The observed interaction of OspE with FH19-20 allows the full function of FH in down-regulation of complement activation on the bacteria. This reveals the molecular basis for how B. burgdorferi evades innate immunity and suggests how OspE could be used as a potential vaccine antigen.
Collapse
Affiliation(s)
- Arnab Bhattacharjee
- Haartman Institute, Department of Bacteriology and Immunology, and Research Programs Unit, Immunobiology, University of Helsinki, FIN-00014 Helsinki, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
247
|
Inoue Y, Takayanagi M, Sugiyama H. Presynaptic protein synaptotagmin1 regulates the activity-induced remodeling of synaptic structures in cultured hippocampal neurons. J Neurosci Res 2013; 91:882-9. [PMID: 23606502 DOI: 10.1002/jnr.23215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 01/07/2013] [Accepted: 01/13/2013] [Indexed: 11/11/2022]
Abstract
Activity-dependent reorganizations of central neuronal synapses are thought to play important roles in learning and memory. Although the precise mechanisms of how neuronal activities modify synaptic connections in neurons remain to be clarified, the activity-induced neuronal presynaptic proteins such as synaptotagmin1 may contribute to the onset of synaptic remodeling. To understand better the physiological roles of synaptotagmin1, we first examined the prolonged effects of neuronal stimulation capable of inducing synaptotagmin1 on the distribution of a postsynaptic proteins (PSD) protein Homer1c by immunostaining. Previously we found that glutamate stimulation induced other postsynaptic proteins, such as postsynaptic density-95 (PSD95), a biphasic change with an initially diffuse distribution after 30 min to 1 hr, followed by reassembly to more than the original level after 4-8 hr, suggesting that glutamate stimulation induces a global biphasic alteration in synaptic structures. To dissect further the functions of synaptotagmin1 in the activity-induced synaptic remodeling, short hairpin RNA (shRNA) vectors that specifically block the expression of endogenous synaptotagmin1 were constructed. When the shRNA of synaptotagmin1 was introduced to the neurons, the activity-induced changes were almost completely suppressed. We found that synaptotagmin1 contributes to the postsynaptic remodeling in a retrograde manner. Our data indicate that synaptotagmin1 regulates the activity-induced biphasic changes of post- and presynaptic sites.
Collapse
Affiliation(s)
- Yuriko Inoue
- Department of Anatomy, School of Medicine, Faculty of Medicine, Toho University, Tokyo, Japan.
| | | | | |
Collapse
|
248
|
Abstract
In recent years, the glutamatergic system has been implicated in the development and treatment of psychiatric disorders. Glutamate signaling is processed by different receptors, including metabotropic glutamate receptors (mGluRs), which in turn interact with the scaffolding protein Homer1 to modulate downstream Ca(2+) signaling. Stress is a major risk factor for the incidence of psychiatric diseases, yet acute stress episodes may have diverging effects on individuals. Cognitive impairments have often been shown to occur after episodes of stress, however the specific role of mGluR5/Homer1 signaling in the interaction of stress and cognition has not yet been elucidated. In this study we show that a single episode of social defeat stress is sufficient to specifically induce cognitive impairments in mice 8 h after the stressor without affecting the animals' locomotion or anxiety levels. We also demonstrate that Homer1b/c levels as well as mGluR5/Homer1b/c interactions in the dorsal hippocampus are reduced up to 8 h after stress. Blockade of mGluR5 during the occurrence of social stress was able to rescue the cognitive impairments. In addition, a specific overexpression of Homer1b/c in the dorsal hippocampus also reversed the behavioral phenotype, indicating that both mGluR5 and Homer1b/c play a crucial role in the mediation of the stress effects. In summary, we could demonstrate that stress induces a cognitive deficit that is likely mediated by mGluR5/Homer1 signaling in the hippocampus. These findings help to reveal the underlying effects of cognitive impairments in patients suffering from stress-related psychiatric disorders.
Collapse
|
249
|
Jardin I, López JJ, Berna-Erro A, Salido GM, Rosado JA. Homer Proteins in Ca2+Entry. IUBMB Life 2013; 65:497-504. [DOI: 10.1002/iub.1162] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Accepted: 02/07/2013] [Indexed: 11/08/2022]
|
250
|
Persistent receptor activity underlies group I mGluR-mediated cellular plasticity in CA3 neuron. J Neurosci 2013; 33:2526-40. [PMID: 23392681 DOI: 10.1523/jneurosci.3338-12.2013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Plastic changes in cortical activities induced by group I metabotropic glutamate receptor (mGluR) stimulation include epileptogenesis, expressed in vitro as the conversion of normal neuronal activity to persistent, prolonged synchronized (ictal) discharges. At present, the mechanism that maintains group I mGluR-induced plasticity is not known. We examined this issue using hippocampal slices from guinea pigs and mice. Agonist [(S)-3,5-dihydroxyphenylglycine (DHPG), 30-50 μm)] stimulation of group I mGluRs induces persistent prolonged synchronized (ictal-like) discharges in CA3 that are associated with three identified excitatory cellular responses-suppression of spike afterhyperpolarizations, activation of a voltage-dependent cationic current, and increase in neuronal input resistance. Persistent prolonged synchronized discharges and the underlying excitatory cellular responses maintained following induction were reversibly blocked by mGluR1 antagonists [(S)-+-α-amino-4-carboxy-2-methylbenzeneacetic acid (LY 367385), 50, 100 μm; CPCCOEt (hydroxyimino)cyclopropa[b]chromen-1a-carboxylate ethyl ester, 100 μm], and to a lesser extent by the mGluR5 antagonist MPEP [2-methyl-6-(phenylethynyl)pyridine hydrochloride, 50 μm]. Activation of persistent cellular responses to DHPG were unaffected by tetrodotoxin (0.5-1 μm) or perfusion with low Ca(2+)(0.2 mm)-Mn(2+)(0.5 mm) media-conditions that suppress endogenous glutamate release. The pharmacological profile of the blocking action of the group I mGluR antagonist MCPG [(RS)-α-methyl-4-carboxyphenylglycine, 50-500 μm] on persistent cellular responses was different from that on cellular responses directly activated by DHPG. These data indicate that transient stimulation of group I mGluRs alters receptor properties, rendering them persistently active in the absence of applied agonist or endogenous glutamate activation. Persistent receptor activities, primarily involving mGluR1, maintain excitatory cellular responses and emergent prolonged synchronized discharges.
Collapse
|