101
|
Wild AR, Dell'Acqua ML. Potential for therapeutic targeting of AKAP signaling complexes in nervous system disorders. Pharmacol Ther 2017; 185:99-121. [PMID: 29262295 DOI: 10.1016/j.pharmthera.2017.12.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
A common feature of neurological and neuropsychiatric disorders is a breakdown in the integrity of intracellular signal transduction pathways. Dysregulation of ion channels and receptors in the cell membrane and the enzymatic mediators that link them to intracellular effectors can lead to synaptic dysfunction and neuronal death. However, therapeutic targeting of these ubiquitous signaling elements can lead to off-target side effects due to their widespread expression in multiple systems of the body. A-kinase anchoring proteins (AKAPs) are multivalent scaffolding proteins that compartmentalize a diverse range of receptor and effector proteins to streamline signaling within nanodomain signalosomes. A number of essential neurological processes are known to critically depend on AKAP-directed signaling and an understanding of the role AKAPs play in nervous system disorders has emerged in recent years. Selective targeting of AKAP protein-protein interactions may be a means to uncouple pathologically active signaling pathways in neurological disorders with a greater degree of specificity. In this review we will discuss the role of AKAPs in both regulating normal nervous system function and dysfunction associated with disease, and the potential for therapeutic targeting of AKAP signaling complexes.
Collapse
Affiliation(s)
- Angela R Wild
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Mark L Dell'Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| |
Collapse
|
102
|
Augustin K, Khabbush A, Williams S, Eaton S, Orford M, Cross JH, Heales SJR, Walker MC, Williams RSB. Mechanisms of action for the medium-chain triglyceride ketogenic diet in neurological and metabolic disorders. Lancet Neurol 2017; 17:84-93. [PMID: 29263011 DOI: 10.1016/s1474-4422(17)30408-8] [Citation(s) in RCA: 268] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 11/03/2017] [Accepted: 11/20/2017] [Indexed: 12/16/2022]
Abstract
High-fat, low-carbohydrate diets, known as ketogenic diets, have been used as a non-pharmacological treatment for refractory epilepsy. A key mechanism of this treatment is thought to be the generation of ketones, which provide brain cells (neurons and astrocytes) with an energy source that is more efficient than glucose, resulting in beneficial downstream metabolic changes, such as increasing adenosine levels, which might have effects on seizure control. However, some studies have challenged the central role of ketones because medium-chain fatty acids, which are part of a commonly used variation of the diet (the medium-chain triglyceride ketogenic diet), have been shown to directly inhibit AMPA receptors (glutamate receptors), and to change cell energetics through mitochondrial biogenesis. Through these mechanisms, medium-chain fatty acids rather than ketones are likely to block seizure onset and raise seizure threshold. The mechanisms underlying the ketogenic diet might also have roles in other disorders, such as preventing neurodegeneration in Alzheimer's disease, the proliferation and spread of cancer, and insulin resistance in type 2 diabetes. Analysing medium-chain fatty acids in future ketogenic diet studies will provide further insights into their importance in modified forms of the diet. Moreover, the results of these studies could facilitate the development of new pharmacological and dietary therapies for epilepsy and other disorders.
Collapse
Affiliation(s)
- Katrin Augustin
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, UK
| | - Aziza Khabbush
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Sophie Williams
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, University College London, London, UK
| | - Simon Eaton
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Michael Orford
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - J Helen Cross
- Neurosciences Unit, UCL Institute of Child Health, University College London, London, UK
| | - Simon J R Heales
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Matthew C Walker
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, University College London, London, UK
| | - Robin S B Williams
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, UK.
| |
Collapse
|
103
|
Wei IH, Chen KT, Tsai MH, Wu CH, Lane HY, Huang CC. Acute Amino Acid d-Serine Administration, Similar to Ketamine, Produces Antidepressant-like Effects through Identical Mechanisms. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:10792-10803. [PMID: 29161812 DOI: 10.1021/acs.jafc.7b04217] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
d-Serine is an amino acid and can work as an agonist at the glycine sites of N-methyl-d-aspartate receptor (NMDAR). Interestingly, both types of glutamatergic modulators, NMDAR enhancers and blockers, can improve depression through common targets, namely alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionaic acid receptors (AMPARs) and mammalian target of rapamycin (mTOR). To elucidate the cellular signaling pathway underlying this counterintuitive observation, we activated NMDARs in rats by using d-serine. Saline, ketamine (NMDAR antagonist), and desipramine (tricyclic antidepressant) were used as controls. The antidepressant-like effects of all agents were evaluated using the forced swim test. The activation of the AMPAR-mTOR signaling pathway, release of brain-derived neurotrophic factor (BDNF), and alteration of AMPAR and NMDAR trafficking in the hippocampus of rats were examined. A single high dose of d-serine exerted an antidepressant-like effect that was mediated by rapid AMPAR-induced mTOR signaling pathway and increased BDNF proteins, identical to that of ketamine. Furthermore, in addition to the increased protein kinase A phosphorylation of the AMPAR subunit GluR1 (an indicator of AMPAR insertion in neurons), treatment with individual optimal doses of d-serine and ketamine also increased adaptin β2-NMDAR association (an indicator of the intracellular endocytic machinery and subsequent internalization of NMDARs). Desipramine did not influence these processes. Our study is the first to demonstrate an association between d-serine and ketamine; following adaptative regulation of AMPAR and NMDAR may lead to common changes of them. These findings provide novel targets for safer antidepressant agents with mechanisms similar to those of ketamine.
Collapse
Affiliation(s)
| | | | | | - Ching-Hsiang Wu
- Department of Anatomy, College of Medicine, Taipei Medical University , 110 Taipei, Taiwan
| | - Hsien-Yuan Lane
- Brain Disease Research Center & Department of Psychiatry, China Medical University Hospital , 404 Taichung, Taiwan
| | - Chih-Chia Huang
- Brain Disease Research Center & Department of Psychiatry, China Medical University Hospital , 404 Taichung, Taiwan
| |
Collapse
|
104
|
Shen Y, Tian M, Zheng Y, Gong F, Fu AKY, Ip NY. Stimulation of the Hippocampal POMC/MC4R Circuit Alleviates Synaptic Plasticity Impairment in an Alzheimer's Disease Model. Cell Rep 2017; 17:1819-1831. [PMID: 27829153 DOI: 10.1016/j.celrep.2016.10.043] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 07/27/2016] [Accepted: 10/13/2016] [Indexed: 02/07/2023] Open
Abstract
Hippocampal synaptic plasticity is modulated by neuropeptides, the disruption of which might contribute to cognitive deficits observed in Alzheimer's disease (AD). Although pro-opiomelanocortin (POMC)-derived neuropeptides and melanocortin 4 receptor (MC4R) are implicated in hippocampus-dependent synaptic plasticity, how the POMC/MC4R system functions in the hippocampus and its role in synaptic dysfunction in AD are largely unknown. Here, we mapped a functional POMC circuit in the mouse hippocampus, wherein POMC neurons in the cornu ammonis 3 (CA3) activate MC4R in the CA1. Suppression of hippocampal MC4R activity in the APP/PS1 transgenic mouse model of AD exacerbates long-term potentiation impairment, which is alleviated by the replenishment of hippocampal POMC/MC4R activity or activation of hippocampal MC4R-coupled Gs signaling. Importantly, MC4R activation rescues amyloid-β-induced synaptic dysfunction via a Gs/cyclic AMP (cAMP)/PKA/cAMP-response element binding protein (CREB)-dependent mechanism. Hence, disruption of this hippocampal POMC/MC4R circuit might contribute to synaptic dysfunction observed in AD, revealing a potential therapeutic target for the disease.
Collapse
Affiliation(s)
- Yang Shen
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Min Tian
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Yuqiong Zheng
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Fei Gong
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Amy K Y Fu
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Nancy Y Ip
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.
| |
Collapse
|
105
|
Enhanced AMPA Receptor Trafficking Mediates the Anorexigenic Effect of Endogenous Glucagon-like Peptide-1 in the Paraventricular Hypothalamus. Neuron 2017; 96:897-909.e5. [PMID: 29056294 DOI: 10.1016/j.neuron.2017.09.042] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 08/16/2017] [Accepted: 09/22/2017] [Indexed: 12/27/2022]
Abstract
Glucagon-like Peptide 1 (GLP-1)-expressing neurons in the hindbrain send robust projections to the paraventricular nucleus of the hypothalamus (PVN), which is involved in the regulation of food intake. Here, we describe that stimulation of GLP-1 afferent fibers within the PVN is sufficient to suppress food intake independent of glutamate release. We also show that GLP-1 receptor (GLP-1R) activation augments excitatory synaptic strength in PVN corticotropin-releasing hormone (CRH) neurons, with GLP-1R activation promoting a protein kinase A (PKA)-dependent signaling cascade leading to phosphorylation of serine S845 on GluA1 AMPA receptors and their trafficking to the plasma membrane. Finally, we show that postnatal depletion of GLP-1R in the PVN increases food intake and causes obesity. This study provides a comprehensive multi-level (circuit, synaptic, and molecular) explanation of how food intake behavior and body weight are regulated by endogenous central GLP-1. VIDEO ABSTRACT.
Collapse
|
106
|
Caffino L, Piva A, Mottarlini F, Di Chio M, Giannotti G, Chiamulera C, Fumagalli F. Ketamine Self-Administration Elevates αCaMKII Autophosphorylation in Mood and Reward-Related Brain Regions in Rats. Mol Neurobiol 2017; 55:5453-5461. [PMID: 28948570 DOI: 10.1007/s12035-017-0772-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 09/12/2017] [Indexed: 12/30/2022]
Abstract
Modulation of αCaMKII expression and phosphorylation is a feature shared by drugs of abuse with different mechanisms of action. Accordingly, we investigated whether αCaMKII expression and activation could be altered by self-administration of ketamine, a non-competitive antagonist of the NMDA glutamate receptor, with antidepressant and psychotomimetic as well as reinforcing properties. Rats self-administered ketamine at a sub-anesthetic dose for 43 days and were sacrificed 24 h after the last drug exposure; reward-related brain regions, such as medial prefrontal cortex (PFC), ventral striatum (vS), and hippocampus (Hip), were used for the measurement of αCaMKII-mediated signaling. αCaMKII phosphorylation was increased in these brain regions suggesting that ketamine, similarly to other reinforcers, activates this kinase. We next measured the two main targets of αCaMKII, i.e., GluN2B (S1303) and GluA1 (S831), and found increased activation of GluN2B (S1303) together with reduced phosphorylation of GluA1 (S831). Since GluN2B, via inhibition of ERK, regulates the membrane expression of GluA1, we measured ERK2 phosphorylation in the crude synaptosomal fraction of these brain regions, which was significantly reduced suggesting that ketamine-induced phosphorylation of αCaMKII promotes GluN2B (S1303) phosphorylation that, in turn, inhibits ERK 2 signaling, an effect that results in reduced membrane expression and phosphorylation of GluA1. Taken together, our findings point to αCaMKII autophosphorylation as a critical signature of ketamine self-administration providing an intracellular mechanism to explain the different effects caused by αCaMKII autophosphorylation on the post-synaptic GluN2B- and GluA1-mediated functions. These data add ketamine to the list of drugs of abuse converging on αCaMKII to sustain their addictive properties.
Collapse
Affiliation(s)
- Lucia Caffino
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Alessandro Piva
- Neuropsychopharmacology Laboratory, Section Pharmacology, Department of Diagnostic and Public Health, P.le Scuro 10, University of Verona, Verona, Italy
| | - Francesca Mottarlini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Marzia Di Chio
- Neuropsychopharmacology Laboratory, Section Pharmacology, Department of Diagnostic and Public Health, P.le Scuro 10, University of Verona, Verona, Italy
| | - Giuseppe Giannotti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Cristiano Chiamulera
- Neuropsychopharmacology Laboratory, Section Pharmacology, Department of Diagnostic and Public Health, P.le Scuro 10, University of Verona, Verona, Italy
| | - Fabio Fumagalli
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy.
| |
Collapse
|
107
|
Liu X, Zhong P, Vickstrom C, Li Y, Liu QS. PDE4 Inhibition Restores the Balance Between Excitation and Inhibition in VTA Dopamine Neurons Disrupted by Repeated In Vivo Cocaine Exposure. Neuropsychopharmacology 2017; 42:1991-1999. [PMID: 28497801 PMCID: PMC5561351 DOI: 10.1038/npp.2017.96] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 04/26/2017] [Accepted: 05/09/2017] [Indexed: 01/01/2023]
Abstract
Phosphodiesterase type 4 (PDE4) is a family of enzymes that selectively degrade intracellular cAMP. PDE4 inhibitors have been shown to regulate the rewarding and reinforcing effects of cocaine, but the underlying mechanisms remain poorly understood. Here we show that pretreatments with the PDE4 inhibitor rolipram attenuated cocaine-induced locomotor sensitization in mice. Repeated cocaine exposure in vivo caused a decrease in inhibitory postsynaptic currents (IPSCs) and an increase in the AMPAR/NMDAR ratio in ventral tegmental area (VTA) dopamine neurons in midbrain slices ex vivo. Cocaine exposure disrupted the balance between excitation and inhibition as shown by an increase in the excitation to inhibition (E/I) ratio. Rolipram pretreatments in vivo prevented cocaine-induced reductions in GABAergic inhibition but did not further increase cocaine-induced potentiation of excitation, leading to the restoration of a balance between excitation and inhibition and normalization of the E/I ratio. In support of this idea, we found that repeated cocaine exposure led to an increase in the single-unit action potential firing rate in vivo in VTA dopamine neurons, which was blocked by rolipram pretreatments. These results suggest that repeated cocaine exposure in vivo disrupts the balance between excitation and inhibition in VTA dopamine neurons, while PDE4 inhibition reestablishes the balance between excitation and inhibition through distinct mechanisms.
Collapse
Affiliation(s)
- Xiaojie Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Peng Zhong
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Casey Vickstrom
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yan Li
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Qing-Song Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA,Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226 USA, Tel: +(414) 955-8877, Fax: +(414) 955-6545, E-mail:
| |
Collapse
|
108
|
Ferrara NC, Cullen PK, Pullins SP, Rotondo EK, Helmstetter FJ. Input from the medial geniculate nucleus modulates amygdala encoding of fear memory discrimination. Learn Mem 2017; 24:414-421. [PMID: 28814467 PMCID: PMC5580525 DOI: 10.1101/lm.044131.116] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 07/05/2017] [Indexed: 12/24/2022]
Abstract
Generalization of fear can involve abnormal responding to cues that signal safety and is common in people diagnosed with post-traumatic stress disorder. Differential auditory fear conditioning can be used as a tool to measure changes in fear discrimination and generalization. Most prior work in this area has focused on elevated amygdala activity as a critical component underlying generalization. The amygdala receives input from auditory cortex as well as the medial geniculate nucleus (MgN) of the thalamus, and these synapses undergo plastic changes in response to fear conditioning and are major contributors to the formation of memory related to both safe and threatening cues. The requirement for MgN protein synthesis during auditory discrimination and generalization, as well as the role of MgN plasticity in amygdala encoding of discrimination or generalization, have not been directly tested. GluR1 and GluR2 containing AMPA receptors are found at synapses throughout the amygdala and their expression is persistently up-regulated after learning. Some of these receptors are postsynaptic to terminals from MgN neurons. We found that protein synthesis-dependent plasticity in MgN is necessary for elevated freezing to both aversive and safe auditory cues, and that this is accompanied by changes in the expressions of AMPA receptor and synaptic scaffolding proteins (e.g., SHANK) at amygdala synapses. This work contributes to understanding the neural mechanisms underlying increased fear to safety signals after stress.
Collapse
Affiliation(s)
- Nicole C Ferrara
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, USA
| | - Patrick K Cullen
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, USA
| | - Shane P Pullins
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, USA
| | - Elena K Rotondo
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, USA
| | - Fred J Helmstetter
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, USA
| |
Collapse
|
109
|
Fiuza M, Rostosky CM, Parkinson GT, Bygrave AM, Halemani N, Baptista M, Milosevic I, Hanley JG. PICK1 regulates AMPA receptor endocytosis via direct interactions with AP2 α-appendage and dynamin. J Cell Biol 2017; 216:3323-3338. [PMID: 28855251 PMCID: PMC5626541 DOI: 10.1083/jcb.201701034] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 06/09/2017] [Accepted: 07/18/2017] [Indexed: 01/01/2023] Open
Abstract
Clathrin-mediated endocytosis (CME) is used to internalize a diverse range of cargo proteins from the cell surface, often in response to specific signals. In neurons, the rapid endocytosis of GluA2-containing AMPA receptors (AMPARs) in response to NMDA receptor (NMDAR) stimulation causes a reduction in synaptic strength and is the central mechanism for long-term depression, which underlies certain forms of learning. The mechanisms that link NMDAR activation to CME of AMPARs remain elusive. PICK1 is a BAR domain protein required for NMDAR-dependent reductions in surface GluA2; however, the molecular mechanisms involved are unclear. In this study, we show that PICK1 makes direct, NMDAR-dependent interactions with the core endocytic proteins AP2 and dynamin. PICK1-AP2 interactions are required for clustering AMPARs at endocytic zones in dendrites in response to NMDAR stimulation and for consequent AMPAR internalization. We further show that PICK1 stimulates dynamin polymerization. We propose that PICK1 is a cargo-specific endocytic accessory protein required for efficient, activity-dependent AMPAR endocytosis.
Collapse
Affiliation(s)
- Maria Fiuza
- Centre for Synaptic Plasticity and School of Biochemistry, University of Bristol, Bristol, England, UK
| | - Christine M Rostosky
- European Neuroscience Institute, University Medical Center Göttingen, Göttingen, Germany
| | - Gabrielle T Parkinson
- Centre for Synaptic Plasticity and School of Biochemistry, University of Bristol, Bristol, England, UK
| | - Alexei M Bygrave
- Centre for Synaptic Plasticity and School of Biochemistry, University of Bristol, Bristol, England, UK
| | - Nagaraj Halemani
- Centre for Synaptic Plasticity and School of Biochemistry, University of Bristol, Bristol, England, UK
| | - Marcio Baptista
- Centre for Synaptic Plasticity and School of Biochemistry, University of Bristol, Bristol, England, UK
| | - Ira Milosevic
- European Neuroscience Institute, University Medical Center Göttingen, Göttingen, Germany
| | - Jonathan G Hanley
- Centre for Synaptic Plasticity and School of Biochemistry, University of Bristol, Bristol, England, UK
| |
Collapse
|
110
|
Protein Kinase C γ Contributes to Central Sensitization in a Rat Model of Chronic Migraine. J Mol Neurosci 2017; 63:131-141. [PMID: 28842814 DOI: 10.1007/s12031-017-0960-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/31/2017] [Indexed: 01/01/2023]
Abstract
Protein kinase C γ (PKCγ) is a critical regulator of central sensitization and is widely recognized to be involved in the pathogenesis of chronic migraine (CM). However, the function of PKCγ in CM remains unknown. This study investigated the role of PKCγ on pathogenesis of CM. We repeated infusions of inflammatory soup (IS) on the intact dura of conscious rats to model recurrent trigeminovascular or dural nociceptor activation assumed to occur in patients with CM. The von Frey test was then used to detect changes in pain threshold. QT-PCR, western blotting, and double immunofluorescence staining were performed to detect the expression and location of PKCγ in the trigeminal nucleus caudalis (TNC) and the expressions of calcitonin gene-related peptide (CGRP), c-Fos, and phosphorylation level of GluR1 subunit at serine 831. Chelerythrine chloride (CHE) and phorbol 12-myristate 13-acetate (PMA) were administrated to investigate the role of PKCγ in central sensitization. We found that repeated infusions of IS induced mechanical allodynia. PKCγ was significantly increased in TNC after CM. Furthermore, inhibition of PKCγ by CHE relieved allodynia and reduced the expression of CGRP and c-Fos. Activation of PKCγ by PMA aggravated allodynia and increased the expression of CGRP and c-Fos. In addition, inhibition of PKCγ reduced the phosphorylation level of GluR1; in contrast, activation of PKCγ increased the phosphorylation level of GluR1. These results suggest PKCγ-induced GluR1 phosphorylation might participate in central sensitization in a rat model of CM. We suggest that PKCγ is a potential therapeutic target for CM.
Collapse
|
111
|
De Mario A, Peggion C, Massimino ML, Viviani F, Castellani A, Giacomello M, Lim D, Bertoli A, Sorgato MC. The prion protein regulates glutamate-mediated Ca 2+ entry and mitochondrial Ca 2+ accumulation in neurons. J Cell Sci 2017; 130:2736-2746. [PMID: 28701513 DOI: 10.1242/jcs.196972] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 07/05/2017] [Indexed: 01/01/2023] Open
Abstract
The cellular prion protein (PrPC) whose conformational misfolding leads to the production of deadly prions, has a still-unclarified cellular function despite decades of intensive research. Following our recent finding that PrPC limits Ca2+ entry via store-operated Ca2+ channels in neurons, we investigated whether the protein could also control the activity of ionotropic glutamate receptors (iGluRs). To this end, we compared local Ca2+ movements in primary cerebellar granule neurons and cortical neurons transduced with genetically encoded Ca2+ probes and expressing, or not expressing, PrPC Our investigation demonstrated that PrPC downregulates Ca2+ entry through each specific agonist-stimulated iGluR and after stimulation by glutamate. We found that, although PrP-knockout (KO) mitochondria were displaced from the plasma membrane, glutamate addition resulted in a higher mitochondrial Ca2+ uptake in PrP-KO neurons than in their PrPC-expressing counterpart. This was because the increased Ca2+ entry through iGluRs in PrP-KO neurons led to a parallel increase in Ca2+-induced Ca2+ release via ryanodine receptor channels. These data thus suggest that PrPC takes part in the cell apparatus controlling Ca2+ homeostasis, and that PrPC is involved in protecting neurons from toxic Ca2+ overloads.
Collapse
Affiliation(s)
- Agnese De Mario
- Department of Biomedical Science, University of Padova, 35131 Padova, Italy
| | - Caterina Peggion
- Department of Biomedical Science, University of Padova, 35131 Padova, Italy
| | - Maria Lina Massimino
- CNR Neuroscience Institute, Department of Biomedical Science, University of Padova, 35131 Padova, Italy
| | - Francesca Viviani
- Department of Biomedical Science, University of Padova, 35131 Padova, Italy
| | - Angela Castellani
- Department of Biomedical Science, University of Padova, 35131 Padova, Italy
| | - Marta Giacomello
- Department of Biology, University of Padova, 35131 Padova, Italy
| | - Dmitry Lim
- Department of Pharmaceutical Science, University of Piemonte Orientale, 28100 Novara, Italy
| | - Alessandro Bertoli
- Department of Biomedical Science, University of Padova, 35131 Padova, Italy
| | - Maria Catia Sorgato
- Department of Biomedical Science, University of Padova, 35131 Padova, Italy .,CNR Neuroscience Institute, Department of Biomedical Science, University of Padova, 35131 Padova, Italy
| |
Collapse
|
112
|
Pinar C, Fontaine CJ, Triviño-Paredes J, Lottenberg CP, Gil-Mohapel J, Christie BR. Revisiting the flip side: Long-term depression of synaptic efficacy in the hippocampus. Neurosci Biobehav Rev 2017. [PMID: 28624435 DOI: 10.1016/j.neubiorev.2017.06.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Synaptic plasticity is widely regarded as a putative biological substrate for learning and memory processes. While both decreases and increases in synaptic strength are seen as playing a role in learning and memory, long-term depression (LTD) of synaptic efficacy has received far less attention than its counterpart long-term potentiation (LTP). Never-the-less, LTD at synapses can play an important role in increasing computational flexibility in neural networks. In addition, like learning and memory processes, the magnitude of LTD can be modulated by factors that include stress and sex hormones, neurotrophic support, learning environments, and age. Examining how these factors modulate hippocampal LTD can provide the means to better elucidate the molecular underpinnings of learning and memory processes. This is in turn will enhance our appreciation of how both increases and decreases in synaptic plasticity can play a role in different neurodevelopmental and neurodegenerative conditions.
Collapse
Affiliation(s)
- Cristina Pinar
- Division of Medical Sciences and UBC Island Medical Program, University of Victoria, Victoria, British Columbia, Canada
| | - Christine J Fontaine
- Division of Medical Sciences and UBC Island Medical Program, University of Victoria, Victoria, British Columbia, Canada
| | - Juan Triviño-Paredes
- Division of Medical Sciences and UBC Island Medical Program, University of Victoria, Victoria, British Columbia, Canada
| | - Carina P Lottenberg
- Division of Medical Sciences and UBC Island Medical Program, University of Victoria, Victoria, British Columbia, Canada; Faculty of Medical Sciences of Santa Casa de São Paulo, Sao Paulo, SP, Brazil
| | - Joana Gil-Mohapel
- Division of Medical Sciences and UBC Island Medical Program, University of Victoria, Victoria, British Columbia, Canada
| | - Brian R Christie
- Division of Medical Sciences and UBC Island Medical Program, University of Victoria, Victoria, British Columbia, Canada.
| |
Collapse
|
113
|
Lameth J, Gervais A, Colin C, Lévêque P, Jay TM, Edeline JM, Mallat M. Acute Neuroinflammation Promotes Cell Responses to 1800 MHz GSM Electromagnetic Fields in the Rat Cerebral Cortex. Neurotox Res 2017; 32:444-459. [PMID: 28578480 DOI: 10.1007/s12640-017-9756-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/16/2017] [Accepted: 05/19/2017] [Indexed: 01/08/2023]
Abstract
Mobile phone communications are conveyed by radiofrequency (RF) electromagnetic fields, including pulse-modulated global system for mobile communications (GSM)-1800 MHz, whose effects on the CNS affected by pathological states remain to be specified. Here, we investigated whether a 2-h head-only exposure to GSM-1800 MHz could impact on a neuroinflammatory reaction triggered by lipopolysaccharide (LPS) in 2-week-old or adult rats. We focused on the cerebral cortex in which the specific absorption rate (SAR) of RF averaged 2.9 W/kg. In developing rats, 24 h after GSM exposure, the levels of cortical interleukin-1ß (IL1ß) or NOX2 NADPH oxidase transcripts were reduced by 50 to 60%, in comparison with sham-exposed animals (SAR = 0), as assessed by RT-qPCR. Adult rats exposed to GSM also showed a 50% reduction in the level of IL1ß mRNA, but they differed from developing rats by the lack of NOX2 gene suppression and by displaying a significant growth response of microglial cell processes imaged in anti-Iba1-stained cortical sections. As neuroinflammation is often associated with changes in excitatory neurotransmission, we evaluated changes in expression and phosphorylation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in the adult cerebral cortex by Western blot analyses. We found that GSM exposure decreased phosphorylation at two residues on the GluA1 AMPAR subunit (serine 831 and 845). The GSM-induced changes in gene expressions, microglia, and GluA1 phosphorylation did not persist 72 h after RF exposure and were not observed in the absence of LPS pretreatment. Together, our data provide evidence that GSM-1800 MHz can modulate CNS cell responses triggered by an acute neuroinflammatory state.
Collapse
Affiliation(s)
- Julie Lameth
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U.1127, CNRS, Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié-Salpêtrière, Bat. ICM, 47 boulevard de l'Hôpital, F-75013, Paris, France
| | - Annie Gervais
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U.1127, CNRS, Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié-Salpêtrière, Bat. ICM, 47 boulevard de l'Hôpital, F-75013, Paris, France
| | - Catherine Colin
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U.1127, CNRS, Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié-Salpêtrière, Bat. ICM, 47 boulevard de l'Hôpital, F-75013, Paris, France
| | - Philippe Lévêque
- Université de Limoges, CNRS, XLIM, UMR 7252, 123 avenue Albert Thomas, F-87000, Limoges, France
| | - Thérèse M Jay
- Physiopathologie des Maladies Psychiatriques, Centre de Psychiatrie et Neurosciences, UMR_S894 INSERM, Université Paris Descartes, 102-108 rue de la Santé, 75014, Paris, France
| | - Jean-Marc Edeline
- Paris Saclay Institute of Neuroscience, Neuro-PSI, UMR 9197 CNRS, Université Paris-Sud, 91405, Orsay cedex, France
| | - Michel Mallat
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U.1127, CNRS, Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié-Salpêtrière, Bat. ICM, 47 boulevard de l'Hôpital, F-75013, Paris, France.
| |
Collapse
|
114
|
Chronic stress-induced dendritic reorganization and abundance of synaptosomal PKA-dependent CP-AMPA receptor in the basolateral amygdala in a mouse model of depression. Biochem Biophys Res Commun 2017; 486:671-678. [DOI: 10.1016/j.bbrc.2017.03.093] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 03/19/2017] [Indexed: 11/24/2022]
|
115
|
Caioli S, Severini C, Ciotti T, Florenzano F, Pimpinella D, Petrocchi Passeri P, Balboni G, Polisca P, Lattanzi R, Nisticò R, Negri L, Zona C. Prokineticin system modulation as a new target to counteract the amyloid beta toxicity induced by glutamatergic alterations in an in vitro model of Alzheimer's disease. Neuropharmacology 2017; 116:82-97. [DOI: 10.1016/j.neuropharm.2016.12.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 11/29/2016] [Accepted: 12/14/2016] [Indexed: 12/28/2022]
|
116
|
Yin P, Xu H, Wang Q, Wang J, Yin L, Xu M, Xie Z, Liu W, Cao X. Overexpression of βCaMKII impairs behavioral flexibility and NMDAR-dependent long-term depression in the dentate gyrus. Neuropharmacology 2017; 116:270-287. [DOI: 10.1016/j.neuropharm.2016.12.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 11/17/2016] [Accepted: 12/15/2016] [Indexed: 12/13/2022]
|
117
|
Jo SY, Jung IH, Yi JH, Choi TJ, Lee S, Jung JW, Yun J, Lee YC, Ryu JH, Kim DH. Ethanol extract of the seed of Zizyphus jujuba var. spinosa potentiates hippocampal synaptic transmission through mitogen-activated protein kinase, adenylyl cyclase, and protein kinase A pathways. JOURNAL OF ETHNOPHARMACOLOGY 2017; 200:16-21. [PMID: 28167293 DOI: 10.1016/j.jep.2017.02.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 01/20/2017] [Accepted: 02/03/2017] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As the seed of Zizyphus jujuba var. spinosa (Bunge) Hu ex H.F. Chow (Rhamnaceae) has been used to sleep disturbances in traditional Chinese and Korean medicine, many previous studies have focused on its sedative effect. AIM OF THE STUDY Recently, we reported the neuroprotective effect of the effect of Z. jujuba var. spinosa. However, its effects on synaptic function have not yet been studied. In this project, we examined the action of ethanol extract of the seed of Z. jujuba var. spinosa (DHP1401) on synaptic transmission in the hippocampus. MATERIALS AND METHODS To investigate the effects of DHP1401, field recordings were conducted using hippocampal slices (400µm). Object recognition test was introduced to examine whether DHP1401 affect normal recognition memory. RESULTS DHP1401 (50μg/ml) induced a significant increase in synaptic activity in Shaffer collateral pathway in a concentration-dependent manner. This increase of synaptic responses was blocked by NBQX, a broad spectrum α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor antagonist, but not IEM-1460, a Ca2+-permeable AMPAR blocker. Moreover, U0126, a mitogen-activated protein kinase inhibitor, SQ22536, an adenylyl cyclase inhibitor, and PKI, a protein kinase A inhibitor, blocked DHP1401-induced increase in synaptic transmission. Finally, DHP1401 facilitated object recognition memory. CONCLUSIONS These results suggest that DHP1401 increase synaptic transmission through increase of synaptic AMPAR transmission via MAPK, AC and PAK.
Collapse
Affiliation(s)
- So Yeon Jo
- Department of Biotechnology, College of Natural Resources and Life Science, Dong-A University, Busan 49315, Republic of Korea.
| | - In Ho Jung
- Department of Life and Nanopharmaceutical Science, Kyung Hee University, Hoeki-dong, Dongdaemoon-Ku, Seoul 02447, Republic of Korea; Daehwa Pharmaceutical Co., Ltd., Seongnam 13488, Republic of Korea.
| | - Jee Hyun Yi
- School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol, Bristol, UK.
| | - Tae Joon Choi
- Daehwa Pharmaceutical Co., Ltd., Seongnam 13488, Republic of Korea.
| | - Seungheon Lee
- Department of Aquatic Biomedical Sciences, School of Marine Biomedical Science, College of Ocean Science, Jeju National University, Jeju 63243, Republic of Korea.
| | - Ji Wook Jung
- Department of Herbal Medicinal Pharmacology, College of Herbal Bio-industry, Daegu Haany University, Kyungsan 38610, Republic of Korea.
| | - Jeanho Yun
- Department of Biochemistry, College of Medicine, Dong-A University, Busan 607-714, Republic of Korea; Institute of Convergence Bio-Health, Dong-A University, Busan 49315, Republic of Korea.
| | - Young Choon Lee
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan 49315, Republic of Korea; Institute of Convergence Bio-Health, Dong-A University, Busan 49315, Republic of Korea.
| | - Jong Hoon Ryu
- Department of Life and Nanopharmaceutical Science, Kyung Hee University, Hoeki-dong, Dongdaemoon-Ku, Seoul 02447, Republic of Korea; Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; Kyung Hee East-West Pharmaceutical Research Institute, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Dong Hyun Kim
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan 49315, Republic of Korea; Institute of Convergence Bio-Health, Dong-A University, Busan 49315, Republic of Korea.
| |
Collapse
|
118
|
Schiff HC, Johansen JP, Hou M, Bush DEA, Smith EK, Klein JE, LeDoux JE, Sears RM. β-Adrenergic Receptors Regulate the Acquisition and Consolidation Phases of Aversive Memory Formation Through Distinct, Temporally Regulated Signaling Pathways. Neuropsychopharmacology 2017; 42:895-903. [PMID: 27762270 PMCID: PMC5312069 DOI: 10.1038/npp.2016.238] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 10/05/2016] [Accepted: 10/07/2016] [Indexed: 01/13/2023]
Abstract
Memory formation requires the temporal coordination of molecular events and cellular processes following a learned event. During Pavlovian threat (fear) conditioning (PTC), sensory and neuromodulatory inputs converge on post-synaptic neurons within the lateral nucleus of the amygdala (LA). By activating an intracellular cascade of signaling molecules, these G-protein-coupled neuromodulatory receptors are capable of recruiting a diverse profile of plasticity-related proteins. Here we report that norepinephrine, through its actions on β-adrenergic receptors (βARs), modulates aversive memory formation following PTC through two molecularly and temporally distinct signaling mechanisms. Specifically, using behavioral pharmacology and biochemistry in adult rats, we determined that βAR activity during, but not after PTC training initiates the activation of two plasticity-related targets: AMPA receptors (AMPARs) for memory acquisition and short-term memory and extracellular regulated kinase (ERK) for consolidating the learned association into a long-term memory. These findings reveal that βAR activity during, but not following PTC sets in motion cascading molecular events for the acquisition (AMPARs) and subsequent consolidation (ERK) of learned associations.
Collapse
Affiliation(s)
- Hillary C Schiff
- Center for Neural Science, New York University, New York, NY, USA
| | - Joshua P Johansen
- RIKEN Brain Science Institute, Laboratory for Neural Circuitry of Memory, Wako-shi, Saitama, Japan
- Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Tokyo, Japan
| | - Mian Hou
- Center for Neural Science, New York University, New York, NY, USA
| | - David E A Bush
- Center for Neural Science, New York University, New York, NY, USA
| | - Emily K Smith
- Center for Neural Science, New York University, New York, NY, USA
| | - JoAnna E Klein
- Center for Neural Science, New York University, New York, NY, USA
| | - Joseph E LeDoux
- Center for Neural Science, New York University, New York, NY, USA
- Department of Psychology, New York University, New York, NY, USA
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Robert M Sears
- Center for Neural Science, New York University, New York, NY, USA
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
- Department of Child and Adolescent Psychiatry, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
119
|
Kugathasan P, Waller J, Westrich L, Abdourahman A, Tamm JA, Pehrson AL, Dale E, Gulinello M, Sanchez C, Li Y. In vivo and in vitro effects of vortioxetine on molecules associated with neuroplasticity. J Psychopharmacol 2017; 31:365-376. [PMID: 27678087 DOI: 10.1177/0269881116667710] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Neuroplasticity is fundamental for brain functions, abnormal changes of which are associated with mood disorders and cognitive impairment. Neuroplasticity can be affected by neuroactive medications and by aging. Vortioxetine, a multimodal antidepressant, has shown positive effects on cognitive functions in both pre-clinical and clinical studies. In rodent studies, vortioxetine increases glutamate neurotransmission, promotes dendritic branching and spine maturation, and elevates hippocampal expression of the activity-regulated cytoskeleton-associated protein (Arc/Arg3.1) at the transcript level. The present study aims to assess the effects of vortioxetine on several neuroplasticity-related molecules in different experimental systems. Chronic (1 month) vortioxetine increased Arc/Arg3.1 protein levels in the cortical synaptosomes of young and middle-aged mice. In young mice, this was accompanied by an increase in actin-depolymerizing factor (ADF)/cofilin serine 3 phosphorylation without altering the total ADF/cofilin protein level, and an increase in the GluA1 subunit of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor phosphorylation at serine 845 (S845) without altering serine 831 (S831) GluA1 phosphorylation nor the total GluA1 protein level. Similar effects were detected in cultured rat hippocampal neurons: Acute vortioxetine increased S845 GluA1 phosphorylation without changing S831 GluA1 phosphorylation or the total GluA1 protein level. These changes were accompanied by an increase in α subunit of Ca2+/calmodulin-dependent kinase (CaMKIIα) phosphorylation (at threonine 286) without changing the total CaMKIIα protein level in cultured neurons. In addition, chronic (1 month) vortioxetine, but not fluoxetine, restored the age-associated reduction in Arc/Arg3.1 and c-Fos transcripts in the frontal cortex of middle-aged mice. Taken together, these results demonstrated that vortioxetine modulates molecular targets that are related to neuroplasticity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yan Li
- 1 Lundbeck Research, Paramus, NJ, USA
| |
Collapse
|
120
|
Ouyang J, Carcea I, Schiavo JK, Jones KT, Rabinowitsch A, Kolaric R, Cabeza de Vaca S, Froemke RC, Carr KD. Food restriction induces synaptic incorporation of calcium-permeable AMPA receptors in nucleus accumbens. Eur J Neurosci 2017; 45:826-836. [PMID: 28112453 PMCID: PMC5359088 DOI: 10.1111/ejn.13528] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 01/14/2017] [Accepted: 01/17/2017] [Indexed: 12/25/2022]
Abstract
Chronic food restriction potentiates behavioral and cellular responses to drugs of abuse and D-1 dopamine receptor agonists administered systemically or locally in the nucleus accumbens (NAc). However, the alterations in NAc synaptic transmission underlying these effects are incompletely understood. AMPA receptor trafficking is a major mechanism for regulating synaptic strength, and previous studies have shown that both sucrose and d-amphetamine rapidly alter the abundance of AMPA receptor subunits in the NAc postsynaptic density (PSD) in a manner that differs between food-restricted and ad libitum fed rats. In this study we examined whether food restriction, in the absence of reward stimulus challenge, alters AMPAR subunit abundance in the NAc PSD. Food restriction was found to increase surface expression and, specifically, PSD abundance, of GluA1 but not GluA2, suggesting synaptic incorporation of GluA2-lacking Ca2+-permeable AMPARs (CP-AMPARs). Naspm, an antagonist of CP-AMPARs, decreased the amplitude of evoked EPSCs in NAc shell, and blocked the enhanced locomotor response to local microinjection of the D-1 receptor agonist, SKF-82958, in food-restricted, but not ad libitum fed, subjects. Although microinjection of the D-2 receptor agonist, quinpirole, also induced greater locomotor activation in food-restricted than ad libitum fed rats, this effect was not decreased by Naspm. Taken together, the present findings are consistent with the synaptic incorporation of CP-AMPARs in D-1 receptor-expressing medium spiny neurons in NAc as a mechanistic underpinning of the enhanced responsiveness of food-restricted rats to natural rewards and drugs of abuse.
Collapse
Affiliation(s)
- Jiangyong Ouyang
- Department of Psychiatry, New York University School of Medicine, 550 First Avenue, New York, New York 10016
| | - Ioana Carcea
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, 550 First Avenue, New York, New York 10016
- Department of Otolaryngology, New York University School of Medicine, 550 First Avenue, New York, New York 10016
- Department of Neuroscience/Physiology, New York University School of Medicine, 550 First Avenue, New York, New York 10016
| | - Jennifer K. Schiavo
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, 550 First Avenue, New York, New York 10016
- Department of Otolaryngology, New York University School of Medicine, 550 First Avenue, New York, New York 10016
- Department of Neuroscience/Physiology, New York University School of Medicine, 550 First Avenue, New York, New York 10016
| | - Kymry T. Jones
- Department of Psychiatry, New York University School of Medicine, 550 First Avenue, New York, New York 10016
| | - Ariana Rabinowitsch
- Department of Psychiatry, New York University School of Medicine, 550 First Avenue, New York, New York 10016
| | - Rhonda Kolaric
- Department of Psychiatry, New York University School of Medicine, 550 First Avenue, New York, New York 10016
| | - Soledad Cabeza de Vaca
- Department of Psychiatry, New York University School of Medicine, 550 First Avenue, New York, New York 10016
| | - Robert C. Froemke
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, 550 First Avenue, New York, New York 10016
- Department of Otolaryngology, New York University School of Medicine, 550 First Avenue, New York, New York 10016
- Department of Neuroscience/Physiology, New York University School of Medicine, 550 First Avenue, New York, New York 10016
| | - Kenneth D. Carr
- Department of Psychiatry, New York University School of Medicine, 550 First Avenue, New York, New York 10016
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, 550 First Avenue, New York, New York 10016
| |
Collapse
|
121
|
Sensitizing exposure to amphetamine increases AMPA receptor phosphorylation without increasing cell surface expression in the rat nucleus accumbens. Neuropharmacology 2017; 117:328-337. [PMID: 28223211 DOI: 10.1016/j.neuropharm.2017.02.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 12/21/2016] [Accepted: 02/17/2017] [Indexed: 12/14/2022]
Abstract
Exposure to psychostimulants like cocaine or amphetamine leads to long-lasting sensitization of their behavioral and neurochemical effects. Here we characterized changes in AMPA receptor distribution and phosphorylation state in the rat nucleus accumbens (NAcc) weeks after amphetamine exposure to assess their potential contribution to sensitization by this drug. Using protein cross-linking, biochemical, subcellular fractionation, and slice electrophysiological approaches in the NAcc, we found that, unlike cocaine, previous exposure to amphetamine did not increase cell surface levels of either GluA1 or GluA2 AMPA receptor subunits, redistribution of these subunits to the synaptic or perisynaptic cellular membrane domains, protein-protein associations required to support the accumulation and retention of AMPA receptors in the PSD, or the peak amplitude of AMPA receptor mediated mEPSCs recorded in NAcc slices. On the other hand, exposure to amphetamine significantly slowed mEPSC decay times and increased levels in the PSD of PKA and CaMKII as well as phosphorylation by these kinases of the GluA1 S845 and S831 residues selectively in this cellular compartment. As the latter effects are known to respectively regulate channel open probability and duration as well as conductance, they provide a novel mechanism that could contribute to the long-lasting AMPA receptor dependent expression of sensitization by amphetamine. Rather than increase the number of surface and synaptic AMPA receptors as with cocaine, this mechanism could increase NAcc medium spiny neuron reactivity to glutamate afferents by increasing the phosphorylation state of critical regulatory sites in the AMPA receptor GluA1 subunit in the PSD.
Collapse
|
122
|
Salas IH, Callaerts-Vegh Z, Arranz AM, Guix FX, D’Hooge R, Esteban JA, De Strooper B, Dotti CG. Tetraspanin 6: A novel regulator of hippocampal synaptic transmission and long term plasticity. PLoS One 2017; 12:e0171968. [PMID: 28207852 PMCID: PMC5312877 DOI: 10.1371/journal.pone.0171968] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 01/27/2017] [Indexed: 11/19/2022] Open
Abstract
Tetraspanins (Tspan) are transmembrane proteins with important scaffold and signalling functions. Deletions of Tetraspanin 6 (Tspan6) gene, a member of the tetraspanin family, have been reported in patients with Epilepsy Female-restricted with Mental Retardation (EFMR). Interestingly, mutations in Tspan7, highly homologous to Tspan6, are associated with X-linked intellectual disability, suggesting that these two proteins are important for cognition. Considering recent evidences showing that Tspan7 plays a key role in synapse development and AMPAR trafficking, we initiated the study of Tspan6 in synaptic function using a Tspan6 knock out mouse model. Here we report that hippocampal field recordings from Tspan6 knock out mice show an enhanced basal synaptic transmission and impaired long term potentiation (LTP). A normal paired-pulse facilitation response suggests that Tspan6 affects the properties of the postsynaptic rather than the presynaptic terminal. However, no changes in spine morphology or postsynaptic markers could be detected in Tspan6 KO mice compared with wild types. In addition, Tspan6 KO mice show normal locomotor behaviour and no defects in hippocampus-dependent memory tests.
Collapse
Affiliation(s)
- Isabel H. Salas
- VIB Center for Biology of Disease – VIB, Leuven, Belgium
- Center of Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND), KU Leuven, Leuven, Gasthuisberg O&N4, Belgium
| | | | - Amaia M. Arranz
- VIB Center for Biology of Disease – VIB, Leuven, Belgium
- Center of Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND), KU Leuven, Leuven, Gasthuisberg O&N4, Belgium
| | - Francesc X. Guix
- VIB Center for Biology of Disease – VIB, Leuven, Belgium
- Center of Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND), KU Leuven, Leuven, Gasthuisberg O&N4, Belgium
| | - Rudi D’Hooge
- Laboratory of Biological Psychology, KU Leuven, Leuven, Belgium
| | - José A. Esteban
- Centro de Biologıa Molecular ‘Severo Ochoa’ (CSIC/UAM), Madrid, Spain
| | - Bart De Strooper
- VIB Center for Biology of Disease – VIB, Leuven, Belgium
- Center of Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND), KU Leuven, Leuven, Gasthuisberg O&N4, Belgium
- * E-mail: (CGD); (BDS)
| | - Carlos G. Dotti
- Centro de Biologıa Molecular ‘Severo Ochoa’ (CSIC/UAM), Madrid, Spain
- * E-mail: (CGD); (BDS)
| |
Collapse
|
123
|
Kulasiri D, Liang J, He Y, Samarasinghe S. Global sensitivity analysis of a model related to memory formation in synapses: Model reduction based on epistemic parameter uncertainties and related issues. J Theor Biol 2017; 419:116-136. [PMID: 28189671 DOI: 10.1016/j.jtbi.2017.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 01/30/2017] [Accepted: 02/04/2017] [Indexed: 11/28/2022]
Abstract
We investigate the epistemic uncertainties of parameters of a mathematical model that describes the dynamics of CaMKII-NMDAR complex related to memory formation in synapses using global sensitivity analysis (GSA). The model, which was published in this journal, is nonlinear and complex with Ca2+ patterns with different level of frequencies as inputs. We explore the effects of parameter on the key outputs of the model to discover the most sensitive ones using GSA and partial ranking correlation coefficient (PRCC) and to understand why they are sensitive and others are not based on the biology of the problem. We also extend the model to add presynaptic neurotransmitter vesicles release to have action potentials as inputs of different frequencies. We perform GSA on this extended model to show that the parameter sensitivities are different for the extended model as shown by PRCC landscapes. Based on the results of GSA and PRCC, we reduce the original model to a less complex model taking the most important biological processes into account. We validate the reduced model against the outputs of the original model. We show that the parameter sensitivities are dependent on the inputs and GSA would make us understand the sensitivities and the importance of the parameters. A thorough phenomenological understanding of the relationships involved is essential to interpret the results of GSA and hence for the possible model reduction.
Collapse
Affiliation(s)
- Don Kulasiri
- Centre for Advanced Computational Solutions (C-fACS), Molecular Biosciences Department, Lincoln University, Christchurch, New Zealand.
| | - Jingyi Liang
- Centre for Advanced Computational Solutions (C-fACS), Molecular Biosciences Department, Lincoln University, Christchurch, New Zealand
| | - Yao He
- Centre for Advanced Computational Solutions (C-fACS), Molecular Biosciences Department, Lincoln University, Christchurch, New Zealand
| | - Sandhya Samarasinghe
- Centre for Advanced Computational Solutions (C-fACS), Molecular Biosciences Department, Lincoln University, Christchurch, New Zealand
| |
Collapse
|
124
|
Li S, Han J, Wang DS, Yang Q, Feng B, Kang WB, Yang L, Liu G, Zhao MG. Sinomenine attenuates chronic inflammatory pain in mice. Metab Brain Dis 2017; 32:211-219. [PMID: 27585465 DOI: 10.1007/s11011-016-9889-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 08/01/2016] [Indexed: 01/27/2023]
Abstract
Sinomenine, an alkaloid originally isolated from the roots of Sinomeniumacutum, is used as a traditional Chinese medicine for rheumatic arthritis. However, little is known about the neuronal mechanisms underlying the analgesic effects of sinomenine in animals with chronic inflammatory pain. In this study, we investigated the persistent inflammatory pain induced by hind paw injection of complete Freund's adjuvant (CFA) in mice, which was reversed by sinomenine administration. In the anterior cingulate cortex (ACC), a region highly associated with chronic pain processing, the upregulation of GluN2B-containing N-methyl-D-aspartate (NMDA) receptors and Ca2+/calmodulin-dependent protein kinase II, total levels of GluA1, and phosphorylation of GluA1 at Ser831 (p-GluA1-Ser831) were reversed by systemically administrating sinomenine. Furthermore, sinomenine treatment downregulated the mammalian target of rapamycin (mTOR) pathway. Increases in p-mTOR, p-p70S6k, p-S6, and p-4EBP, which were induced by chronic inflammation, were all changed. However, sinomenine did not affect the levels of GluN2A-containing NMDA receptors and p-GluA1-Ser845, as well as the total levels of mTOR, p70S6k, S6, and 4EBP. In conclusion, results indicated that sinomenine reduced the chronic inflammatory pain induced by CFA, at least partially by regulating the GluN2B receptors and mTOR signals in the ACC.
Collapse
Affiliation(s)
- Shuo Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Jing Han
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Dong-Sheng Wang
- Jinling Hospital, Department of Orthopedics, Nanjing University, School of Medicine, Nanjing, 210002, China
| | - Qi Yang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Bin Feng
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Wen-Bo Kang
- Jinling Hospital, Department of Orthopedics, Nanjing University, School of Medicine, Nanjing, 210002, China
| | - Le Yang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Gang Liu
- Jinling Hospital, Department of Orthopedics, Nanjing University, School of Medicine, Nanjing, 210002, China.
| | - Ming-Gao Zhao
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
125
|
Sinnen BL, Bowen AB, Forte JS, Hiester BG, Crosby KC, Gibson ES, Dell'Acqua ML, Kennedy MJ. Optogenetic Control of Synaptic Composition and Function. Neuron 2017; 93:646-660.e5. [PMID: 28132827 DOI: 10.1016/j.neuron.2016.12.037] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 10/22/2016] [Accepted: 12/22/2016] [Indexed: 10/20/2022]
Abstract
The molecular composition of the postsynaptic membrane is sculpted by synaptic activity. During synaptic plasticity at excitatory synapses, numerous structural, signaling, and receptor molecules concentrate at the postsynaptic density (PSD) to regulate synaptic strength. We developed an approach that uses light to tune the abundance of specific molecules in the PSD. We used this approach to investigate the relationship between the number of AMPA-type glutamate receptors in the PSD and synaptic strength. Surprisingly, adding more AMPA receptors to excitatory contacts had little effect on synaptic strength. Instead, we observed increased excitatory input through the apparent addition of new functional sites. Our data support a model where adding AMPA receptors is sufficient to activate synapses that had few receptors to begin with, but that additional remodeling events are required to strengthen established synapses. More broadly, this approach introduces the precise spatiotemporal control of optogenetics to the molecular control of synaptic function.
Collapse
Affiliation(s)
- Brooke L Sinnen
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Aaron B Bowen
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Jeffrey S Forte
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Brian G Hiester
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Kevin C Crosby
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Emily S Gibson
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Mark L Dell'Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Matthew J Kennedy
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| |
Collapse
|
126
|
Qian H, Patriarchi T, Price JL, Matt L, Lee B, Nieves-Cintrón M, Buonarati OR, Chowdhury D, Nanou E, Nystoriak MA, Catterall WA, Poomvanicha M, Hofmann F, Navedo MF, Hell JW. Phosphorylation of Ser1928 mediates the enhanced activity of the L-type Ca2+ channel Cav1.2 by the β2-adrenergic receptor in neurons. Sci Signal 2017; 10:10/463/eaaf9659. [PMID: 28119465 DOI: 10.1126/scisignal.aaf9659] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The L-type Ca2+ channel Cav1.2 controls multiple functions throughout the body including heart rate and neuronal excitability. It is a key mediator of fight-or-flight stress responses triggered by a signaling pathway involving β-adrenergic receptors (βARs), cyclic adenosine monophosphate (cAMP), and protein kinase A (PKA). PKA readily phosphorylates Ser1928 in Cav1.2 in vitro and in vivo, including in rodents and humans. However, S1928A knock-in (KI) mice have normal PKA-mediated L-type channel regulation in the heart, indicating that Ser1928 is not required for regulation of cardiac Cav1.2 by PKA in this tissue. We report that augmentation of L-type currents by PKA in neurons was absent in S1928A KI mice. Furthermore, S1928A KI mice failed to induce long-term potentiation in response to prolonged theta-tetanus (PTT-LTP), a form of synaptic plasticity that requires Cav1.2 and enhancement of its activity by the β2-adrenergic receptor (β2AR)-cAMP-PKA cascade. Thus, there is an unexpected dichotomy in the control of Cav1.2 by PKA in cardiomyocytes and hippocampal neurons.
Collapse
Affiliation(s)
- Hai Qian
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Tommaso Patriarchi
- Department of Pharmacology, University of California, Davis, CA 95616-8636, USA
| | - Jennifer L Price
- Department of Pharmacology, University of California, Davis, CA 95616-8636, USA
| | - Lucas Matt
- Department of Pharmacology, University of California, Davis, CA 95616-8636, USA
| | - Boram Lee
- Department of Pharmacology, University of California, Davis, CA 95616-8636, USA
| | | | - Olivia R Buonarati
- Department of Pharmacology, University of California, Davis, CA 95616-8636, USA
| | | | - Evanthia Nanou
- Department of Pharmacology, University of Washington, Seattle, WA 98195-7280, USA
| | - Matthew A Nystoriak
- Department of Pharmacology, University of California, Davis, CA 95616-8636, USA
| | - William A Catterall
- Department of Pharmacology, University of Washington, Seattle, WA 98195-7280, USA
| | - Montatip Poomvanicha
- Department of Pharmacology and Toxicology, Technical University of Munich, D-80802 Munich, Germany
| | - Franz Hofmann
- Department of Pharmacology and Toxicology, Technical University of Munich, D-80802 Munich, Germany
| | - Manuel F Navedo
- Department of Pharmacology, University of California, Davis, CA 95616-8636, USA.
| | - Johannes W Hell
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242-1109, USA. .,Department of Pharmacology, University of California, Davis, CA 95616-8636, USA
| |
Collapse
|
127
|
Prouty EW, Waterhouse BD, Chandler DJ. Corticotropin releasing factor dose-dependently modulates excitatory synaptic transmission in the noradrenergic nucleus locus coeruleus. Eur J Neurosci 2017; 45:712-722. [PMID: 27973694 DOI: 10.1111/ejn.13501] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 12/05/2016] [Accepted: 12/06/2016] [Indexed: 01/19/2023]
Abstract
The noradrenergic nucleus locus coeruleus (LC) is critically involved in the stress response and receives afferent input from a number of corticotropin releasing factor (CRF) containing structures. Several in vivo and in vitro studies in rat have shown that CRF robustly increases the firing rate of LC neurons in a dose-dependent manner. While it is known that these increases are dependent on CRF receptor subtype 1 and mediated by effects of cAMP intracellular signaling cascades on potassium conductance, the impact of CRF on synaptic transmission within LC has not been clarified. In the present study, we used whole-cell patch clamp electrophysiology to assess how varying concentrations of bath-applied CRF affect AMPA-receptor dependent spontaneous excitatory post-synaptic currents (sEPSCs). Compared to vehicle, 10, 25, and 100 nm CRF had no significant effects on any sEPSC parameters. Fifty nanomolar CRF, however, significantly increased sEPSC amplitude, half-width, and charge transfer, while these measures were significantly decreased by 200 nm CRF. These observations suggest that stress may differentially affect ongoing excitatory synaptic transmission in LC depending on how much CRF is released from presynaptic terminals. Combined with the well-documented effects of CRF on membrane properties and spontaneous LC discharge, these observations may help explain how stress and CRF release are able to modulate the signal to noise ratio of LC neurons. These findings have implications for how stress affects the fidelity of signal transmission and information flow through LC and how it might impact norepinephrine release in the CNS.
Collapse
Affiliation(s)
- Eric W Prouty
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Barry D Waterhouse
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA
| | - Daniel J Chandler
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, 2 Medical Center Drive, Stratford, NJ, 08084, USA
| |
Collapse
|
128
|
Bolger GB. The PDE4 cAMP-Specific Phosphodiesterases: Targets for Drugs with Antidepressant and Memory-Enhancing Action. ADVANCES IN NEUROBIOLOGY 2017; 17:63-102. [PMID: 28956330 DOI: 10.1007/978-3-319-58811-7_4] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The PDE4 cyclic nucleotide phosphodiesterases are essential regulators of cAMP abundance in the CNS through their ability to regulate PKA activity, the phosphorylation of CREB, and other important elements of signal transduction. In pre-clinical models and in early-stage clinical trials, PDE4 inhibitors have been shown to have antidepressant and memory-enhancing activity. However, the development of clinically-useful PDE4 inhibitors for CNS disorders has been limited by variable efficacy and significant side effects. Recent structural studies have greatly enhanced our understanding of the molecular configuration of PDE4 enzymes, especially the "long" PDE4 isoforms that are abundant in the CNS. The new structural data provide a rationale for the development of a new generation of PDE4 inhibitors that specifically act on long PDE4 isoforms. These next generation PDE4 inhibitors may also be capable of targeting the interactions of select long forms with their "partner" proteins, such as RACK1, β-arrestin, and DISC1. They would therefore have the ability to affect cAMP levels in specific cellular compartments and target localized cellular functions, such as synaptic plasticity. These new agents might also be able to target PDE4 populations in select regions of the CNS that are implicated in learning and memory, affect, and cognition. Potential therapeutic uses of these agents could include affective disorders, memory enhancement, and neurogenesis.
Collapse
Affiliation(s)
- Graeme B Bolger
- Departments of Medicine and Pharmacology, University of Alabama at Birmingham, 1720 2nd Avenue South, NP 2501, Birmingham, AL, 35294-3300, USA.
| |
Collapse
|
129
|
Chesworth R, Corbit LH. Recent developments in the behavioural and pharmacological enhancement of extinction of drug seeking. Addict Biol 2017; 22:3-43. [PMID: 26687226 DOI: 10.1111/adb.12337] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 09/13/2015] [Accepted: 10/28/2015] [Indexed: 01/17/2023]
Abstract
One of the principal barriers to overcoming addiction is the propensity to relapse, even after months or years of abstinence. Relapse can be precipitated by cues and contexts associated with drug use; thus, decreasing the conditioned properties of these cues and contexts may assist in preventing relapse. The predictive power of drug cues and contexts can be reduced by repeatedly presenting them in the absence of the drug reinforcer, a process known as extinction. The potential of extinction to limit relapse has generated considerable interest and research over the past few decades. While pre-clinical animal models suggest extinction learning assists relapse prevention, treatment efficacy is often lacking when extinction learning principles are translated into clinical trials. Conklin and Tiffany (Addiction, 2002) suggest the lack of efficacy in clinical practice may be due to limited translation of procedures demonstrated through animal research and propose several methodological improvements to enhance extinction learning for drug addiction. This review will examine recent advances in the behavioural and pharmacological manipulation of extinction learning, based on research from pre-clinical models. In addition, the translation of pre-clinical findings-both those suggested by Conklin and Tiffany () and novel demonstrations from the past 13 years-into clinical trials and the efficacy of these methods in reducing craving and relapse, where available, will be discussed. Finally, we highlight areas where promising pre-clinical models have not yet been integrated into current clinical practice but, if applied, could improve upon existing behavioural and pharmacological methods.
Collapse
|
130
|
Krawczyk MC, Fernández RS, Pedreira ME, Boccia MM. Toward a better understanding on the role of prediction error on memory processes: From bench to clinic. Neurobiol Learn Mem 2016; 142:13-20. [PMID: 28017817 DOI: 10.1016/j.nlm.2016.12.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/14/2016] [Accepted: 12/16/2016] [Indexed: 10/20/2022]
Abstract
Experimental psychology defines Prediction Error (PE) as a mismatch between expected and current events. It represents a unifier concept within the memory field, as it is the driving force of memory acquisition and updating. Prediction error induces updating of consolidated memories in strength or content by memory reconsolidation. This process has two different neurobiological phases, which involves the destabilization (labilization) of a consolidated memory followed by its restabilization. The aim of this work is to emphasize the functional role of PE on the neurobiology of learning and memory, integrating and discussing different research areas: behavioral, neurobiological, computational and clinical psychiatry.
Collapse
Affiliation(s)
- María C Krawczyk
- Laboratorio de Neurofarmacología de los Procesos de Memoria, Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, 5(to)piso, Buenos Aires, Argentina.
| | - Rodrigo S Fernández
- Laboratorio de Neurobiología de la Memoria, Departamento de Fisiología y Biología Molecular y Celular, IFIBYNE-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina. Pab. II, 2(do)piso, Buenos Aires, Argentina.
| | - María E Pedreira
- Laboratorio de Neurobiología de la Memoria, Departamento de Fisiología y Biología Molecular y Celular, IFIBYNE-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina. Pab. II, 2(do)piso, Buenos Aires, Argentina.
| | - Mariano M Boccia
- Laboratorio de Neurofarmacología de los Procesos de Memoria, Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, 5(to)piso, Buenos Aires, Argentina.
| |
Collapse
|
131
|
Bennett MR, Arnold J, Hatton SN, Lagopoulos J. Regulation of fear extinction by long-term depression: The roles of endocannabinoids and brain derived neurotrophic factor. Behav Brain Res 2016; 319:148-164. [PMID: 27867101 DOI: 10.1016/j.bbr.2016.11.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 11/10/2016] [Accepted: 11/15/2016] [Indexed: 11/19/2022]
Abstract
The extinction of a conditioned fear response is of great interest in the search for a means of ameliorating adverse neurobiological changes resulting from stress. The discovery that endocannibinoid (EC) levels are inversely related to the extent of such stress, and that the amygdala is a primary site mediating stress, suggests that ECs in this brain region might play a major role in extinction. Supporting this are the observations that the basolateral complex of the amygdala shows an increase in ECs only during extinction and that early clinical trials indicate that cannabinoid-like agents, when taken orally by patients suffering from post traumatic stress disorder (PTSD), reduce insomnia and nightmares. In order to optimize the potential of these agents to ameliorate symptoms of PTSD four important questions need to be answered: first, what is the identity of the cells that release ECs in the amygdala during extinction; second, what are their sites of action; third, what roles do the ECs play in the alleviation of long- depression (LTD), a process central to extinction; and finally, to what extent does brain derived neurotrophic factor (BDNF) facilitate the release of ECs? A review of the relevant literature is presented in an attempt to answer these questions. It is suggested that the principal cell involved in EC synthesis and release during extinction is the so-called excitatory extinction neuron in the basal nucleus of the amygdala. Furthermore that the main site of action of the ECs is the adjacent calcitonin gene-related peptide inhibitory interneurons, whose normal role of blocking the excitatory neurons is greatly diminished. The molecular pathways leading (during extinction trials) to the synthesis and release of ECs from synaptic spines of extinction neurons, that is potentiated by BDNF, are also delineated in this review. Finally, consideration is given to how the autocrine action of BDNF, linked to the release of ECs, can lead to the sustained release of these, so maintaining extinction over long times.
Collapse
Affiliation(s)
- Maxwell R Bennett
- The Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia.
| | - Jonathon Arnold
- The Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia
| | - Sean N Hatton
- The Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia
| | - Jim Lagopoulos
- The Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia; The Sunshine Coast Mind and Neuroscience, Thompson Institute, The University of the Sunshine Coast, QLD, Australia
| |
Collapse
|
132
|
Modification of Hypoxic Respiratory Response by Protein Tyrosine Kinase in Brainstem Ventral Respiratory Neuron Group. PLoS One 2016; 11:e0165895. [PMID: 27798679 PMCID: PMC5087851 DOI: 10.1371/journal.pone.0165895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 10/19/2016] [Indexed: 12/04/2022] Open
Abstract
Protein tyrosine kinase (PTK) mediated the tyrosine phosphorylation modification of neuronal receptors and ion channels. Whether such modification resulted in changes of physiological functions was not sufficiently studied. In this study we examined whether the hypoxic respiratory response—which is the enhancement of breathing in hypoxic environment could be affected by the inhibition of PTK at brainstem ventral respiratory neuron column (VRC). Experiments were performed on urethane anesthetized adult rabbits. Phrenic nerve discharge was recorded as the central respiratory motor output. Hypoxic respiratory response was produced by ventilating the rabbit with 10% O2-balance 90% N2 for 5 minutes. The responses of phrenic nerve discharge to hypoxia were observed before and after microinjecting PTK inhibitor genistein, AMPA receptor antagonist CNQX, or inactive PTK inhibitor analogue daidzein at the region of ambiguus nucleus (NA) at levels 0–2 mm rostral to obex where the inspiratory subgroup of VRC were recorded. Results were as follows: 1. the hypoxic respiratory response was significantly attenuated after microinjection of genistein and/or CNQX, and no additive effect (i.e., further attenuation of hypoxic respiratory response) was observed when genistein and CNQX were microinjected one after another at the same injection site. Microinjection of daidzein had no effect on hypoxic respiratory response. 2. Fluorescent immunostaining showed that hypoxia significantly increased the number of phosphotyrosine immunopositive neurons in areas surrounding NA and most of these neurons were also immunopositive to glutamate AMPA receptor subunit GluR1. These results suggested that PTK played an important role in regulating the hypoxic respiratory response, possibly through the tyrosine phosphorylation modification of glutamate AMPA receptors on the respiratory neurons of ventral respiratory neuron column.
Collapse
|
133
|
Lentinan produces a robust antidepressant-like effect via enhancing the prefrontal Dectin-1/AMPA receptor signaling pathway. Behav Brain Res 2016; 317:263-271. [PMID: 27693847 DOI: 10.1016/j.bbr.2016.09.062] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 09/24/2016] [Accepted: 09/27/2016] [Indexed: 12/22/2022]
Abstract
Lentinan (LNT) is an immune regulator and its potential and mechanism for the treatment of mood disorder is of our interest. Dectin-1 is a β-glucan (including LNT) receptor that regulates immune functions in many immune cell types. Cumulative evidence has suggested that the glutamatergic system seems to play an important role in the treatment of depression. Here, we studied the antidepressant-like effects of LNT and its therapeutical target in regulating the functions of AMPA receptors. We found that 60min treatment with LNT leads to a significant antidepressant-like effect in the tail suspension test (TST) and the forced swim test (FST) in mice. The antidepressant-like effects of LNT in TST and FST remained after 1day or 5days of injections. Additionally, LNT did not show a hyperactive effect in the open field test. Dectin-1 receptor levels were increased after LNT treatment for 5days and the specific Dectin-1 inhibitor laminarin was able to block the antidepressant-like effects of LNT. After 5days of treatment, LNT enhanced p-GluR1 (S845) in the prefrontal cortex (PFC); however, the total GluR1, GluR2, and GluR3 expression levels remained unchanged. We also found that the AMPA-specific blocker GYKI 52466 was able to block the antidepressant-like effects of LNT. This study identified LNT as a novel antidepressant with clinical potential and a new antidepressant mechanism for regulating prefrontal Dectin-1/AMPA receptor signaling.
Collapse
|
134
|
Zhang K, Yamaki VN, Wei Z, Zheng Y, Cai X. Differential regulation of GluA1 expression by ketamine and memantine. Behav Brain Res 2016; 316:152-159. [PMID: 27599619 DOI: 10.1016/j.bbr.2016.09.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 08/29/2016] [Accepted: 09/01/2016] [Indexed: 12/20/2022]
Abstract
Evidence from preclinical and clinical studies shows that ketamine, a noncompetitive NMDA receptor antagonist, exerts rapid and sustained antidepressant responses. However, ketamine's psychotomimetic side effects and abuse liability limit the clinical use of the compound. Interestingly, memantine, another NMDA receptor channel blocker, processes no defined antidepressant property but is much safer and clinical tolerated. Understanding why ketamine but not memantine exhibits rapid antidepressant responses is important to elucidate the cellular signaling underlying the fast antidepressant actions of ketamine and to design a new safer generation of fast-acting antidepressants. Here we show that ketamine but memantine caused a rapid and sustained antidepressant-like responses in forced swim test (FST). Both drugs enhanced GluA1 S845 phosphorylation and potentiated Schaffer collateral-CA1 synaptic transmission. However, ketamine but not memantine elevated the expression of GluA1. Incubating acutely prepared hippocampal slices with ketamine but not memantine enhanced mTOR phosphorylation in a time course parallel to the time course of GluA1 elevation. Our results suggest that distinct properties in regulation of mTOR phosphorylation and synaptic protein expression may underlie the differential effectiveness of ketamine and memantine in their antidepressant responses.
Collapse
Affiliation(s)
- Ke Zhang
- Department of Physiology, Southern Illinois University School of Medicine, 1135 Lincoln Drive, Carbondale, IL 62901, USA
| | - Vitor Nagai Yamaki
- Department of Physiology, Southern Illinois University School of Medicine, 1135 Lincoln Drive, Carbondale, IL 62901, USA
| | - Zhisheng Wei
- The Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang Road, Guangzhou, Guangdong, 51030, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang Road, Guangzhou, Guangdong 51030, China
| | - Yu Zheng
- The Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang Road, Guangzhou, Guangdong, 51030, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang Road, Guangzhou, Guangdong 51030, China
| | - Xiang Cai
- Department of Physiology, Southern Illinois University School of Medicine, 1135 Lincoln Drive, Carbondale, IL 62901, USA; Center for Integrated Research in Cognitive & Neural Sciences, Southern Illinois University School of Medicine, Carbondale, IL 62901, USA.
| |
Collapse
|
135
|
Chen KT, Wu CH, Tsai MH, Wu YC, Jou MJ, Huang CC, Wei IH. Antidepressant-like effects of long-term sarcosine treatment in rats with or without chronic unpredictable stress. Behav Brain Res 2016; 316:1-10. [PMID: 27555541 DOI: 10.1016/j.bbr.2016.06.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 05/31/2016] [Accepted: 06/02/2016] [Indexed: 01/10/2023]
Abstract
Sarcosine, an N-methyl-d-aspartate receptor enhancer, can improve depression-like behavior in rodent models and depression in humans. We found that a single dose of sarcosine exerted antidepressant-like effects with rapid concomitant increases in the mammalian target of rapamycin (mTOR) signaling pathway activation and enhancement of α-amino-3-hydroxy-5-methylisoxazole-4-propionate receptor (AMPAR) membrane insertion. Sarcosine may play a crucial role in developing novel therapy for depression. For a detailed understanding of sarcosine, this study examined the effects of long-term sarcosine treatment on the forced swim test (FST), mTOR signaling, and AMPAR membrane insertion in rats. The effects of long-term sarcosine treatment were examined in naive rats and rats exposed to chronic unpredictable stress (CUS). Long-term sarcosine treatment (560mg/kg/d for 21 d) significantly ameliorated the increased immobility induced by CUS in the FST, reaffirming the potential role of sarcosine as an antidepressant for depressed patients. The same long-term treatment exhibited no such effect in naive rats despite increased mTOR activation and AMPAR membrane insertion in both groups. Our findings clearly show CUS-exposed rats are sensitive to long-term sarcosine treatment in FST and the response at the same dose is absent in naïve rats. Nevertheless, the distinct sensitivity to long-term sarcosine treatment in rats with or without CUS is not associated with the activated mTOR signaling pathway or increased AMPAR membrane insertion. Additionally, understanding the behavioral and molecular basis of distinct responses is vital important for developing personalized treatment programs to increase the probability of success when treating depression.
Collapse
Affiliation(s)
- Kuang-Ti Chen
- Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Ching-Hsiang Wu
- Department of Anatomy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Mang-Hung Tsai
- Department of Anatomy, China Medical University, Taichung, Taiwan
| | - Ya-Chieh Wu
- Department of Nursing, Ching-Kuo Institute of Management and Health, Keelung, Taiwan
| | - Ming-Jia Jou
- School of Chinese medicine for post baccalaureate, I Shou University, Kaohsiung, Taiwan
| | - Chih-Chia Huang
- Department of Psychiatry, China Medical University Hospital, Taichung, Taiwan; Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan; Department of Psychiatry, China Medical University, Taichung, Taiwan.
| | - I-Hua Wei
- Department of Anatomy, China Medical University, Taichung, Taiwan.
| |
Collapse
|
136
|
Yap KAF, Shetty MS, Garcia-Alvarez G, Lu B, Alagappan D, Oh-Hora M, Sajikumar S, Fivaz M. STIM2 regulates AMPA receptor trafficking and plasticity at hippocampal synapses. Neurobiol Learn Mem 2016; 138:54-61. [PMID: 27544849 DOI: 10.1016/j.nlm.2016.08.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 08/14/2016] [Accepted: 08/16/2016] [Indexed: 10/21/2022]
Abstract
STIM2 is an integral membrane protein of the endoplasmic reticulum (ER) that regulates the activity of plasma membrane (PM) channels at ER-PM contact sites. Recent studies show that STIM2 promotes spine maturation and surface expression of the AMPA receptor (AMPAR) subunit GluA1, hinting at a probable role in synaptic plasticity. Here, we used a Stim2 cKO mouse to explore the function of STIM2 in Long-Term Potentiation (LTP) and Depression (LTD), two widely-studied models of synaptic plasticity implicated in information storage. We found that STIM2 is required for the stable expression of both LTP and LTD at CA3-CA1 hippocampal synapses. Altered plasticity in Stim2 cKO mice is associated with subtle alterations in the shape and density of dendritic spines in CA1 neurons. Further, surface delivery of GluA1 in response to LTP-inducing chemical manipulations was markedly reduced in excitatory neurons derived from Stim2 cKO mice. GluA1 endocytosis following chemically-induced LTD was also impaired in Stim2 cKO neurons. We conclude that STIM2 facilitates synaptic delivery and removal of AMPARs and regulates activity-dependent changes in synaptic strength through a unique mode of communication between the ER and the synapse.
Collapse
Affiliation(s)
- Kenrick An Fu Yap
- Duke-NUS Medical School, programme in Neuroscience and Behavioral Disorders, Singapore 169857, Singapore
| | - Mahesh Shivarama Shetty
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Gisela Garcia-Alvarez
- Duke-NUS Medical School, programme in Neuroscience and Behavioral Disorders, Singapore 169857, Singapore
| | - Bo Lu
- Duke-NUS Medical School, programme in Neuroscience and Behavioral Disorders, Singapore 169857, Singapore
| | - Durgadevi Alagappan
- Duke-NUS Medical School, programme in Neuroscience and Behavioral Disorders, Singapore 169857, Singapore
| | - Masatsugu Oh-Hora
- Division of Molecular Immunology, Medical Institute of Bioregulation, Kyushu University, Japan
| | - Sreedharan Sajikumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Marc Fivaz
- Duke-NUS Medical School, programme in Neuroscience and Behavioral Disorders, Singapore 169857, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
137
|
Yu YJ, Huang CH, Chang CH, Gean PW. Involvement of protein phosphatases in the destabilization of methamphetamine-associated contextual memory. ACTA ACUST UNITED AC 2016; 23:486-93. [PMID: 27531839 PMCID: PMC4986857 DOI: 10.1101/lm.039941.115] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 07/08/2016] [Indexed: 01/20/2023]
Abstract
Destabilization refers to a memory that becomes unstable when reactivated and is susceptible to disruption by amnestic agents. Here we delineated the cellular mechanism underlying the destabilization of drug memory. Mice were conditioned with methamphetamine (MeAM) for 3 d, and drug memory was assessed with a conditioned place preference (CPP) protocol. Anisomycin (ANI) was administered 60 min after the CPP retrieval to disrupt reconsolidation. We found that destabilization of MeAM CPP after the application of ANI was blocked by the N-methyl-d-aspartate receptor (NMDAR) antagonist MK-801 and the NR2B antagonist ifenprodil (IFN) but not by the NR2A antagonist NVP-AAM077 (NVP). In addition, decrease in the phosphorylation of GluR1 at Serine845 (p-GluR1-Ser845), decrease in spine density, and a reduction in the AMPAR/NMDAR ratio in the basolateral amygdala (BLA) were reversed after the MK-801 treatment. The effect of ANI on destabilization was prevented by the protein phosphatase 2B (calcineurin, CaN) inhibitors cyclosporine A (CsA) and FK-506 and the protein phosphatase 1 (PP1) inhibitors calyculin A (CA) and okadaic acid (OA). These results suggest that memory destabilization involves the activation of NR2B-containing NMDARs, which in turn allows the influx of Ca2+. Increased intracellular Ca2+ stimulates CaN, leading to the dephosphorylation and inactivation of inhibitor 1 and the activation of PP1. PP1 then dephosphorylates p-GluR1-Ser845 to elicit AMPA receptor (AMPAR) endocytosis and destabilization of the drug memory.
Collapse
Affiliation(s)
- Yang-Jung Yu
- Institute of Basic Medical Sciences and Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan, Taiwan 701
| | - Chien-Hsuan Huang
- Institute of Basic Medical Sciences and Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan, Taiwan 701
| | - Chih-Hua Chang
- Institute of Basic Medical Sciences and Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan, Taiwan 701
| | - Po-Wu Gean
- Institute of Basic Medical Sciences and Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan, Taiwan 701
| |
Collapse
|
138
|
De Jesús-Cortés H, Lu Y, Anderson RM, Khan MZ, Nath V, McDaniel L, Lutter M, Radley JJ, Pieper AA, Cui H. Loss of estrogen-related receptor alpha disrupts ventral-striatal synaptic function in female mice. Neuroscience 2016; 329:66-73. [PMID: 27155145 PMCID: PMC8916097 DOI: 10.1016/j.neuroscience.2016.04.054] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 04/09/2016] [Accepted: 04/29/2016] [Indexed: 10/21/2022]
Abstract
Eating disorders (EDs), including anorexia nervosa, bulimia nervosa and binge-ED, are mental illnesses characterized by high morbidity and mortality. While several studies have identified neural deficits in patients with EDs, the cellular and molecular basis of the underlying dysfunction has remained poorly understood. We previously identified a rare missense mutation in the transcription factor estrogen-related receptor alpha (ESRRA) associated with development of EDs. Because ventral-striatal signaling is related to the reward and motivation circuitry thought to underlie EDs, we performed functional and structural analysis of ventral-striatal synapses in Esrra-null mice. Esrra-null female, but not male, mice exhibit altered miniature excitatory postsynaptic currents on medium spiny neurons (MSNs) in the ventral striatum, including increased frequency, increased amplitude, and decreased paired pulse ratio. These electrophysiological measures are associated with structural and molecular changes in synapses of MSNs in the ventral striatum, including fewer pre-synaptic glutamatergic vesicles and enhanced GluR1 function. Neuronal Esrra is thus required for maintaining normal synaptic function in the ventral striatum, which may offer mechanistic insights into the behavioral deficits observed in Esrra-null mice.
Collapse
Affiliation(s)
- Héctor De Jesús-Cortés
- Department of Psychiatry, University of Iowa, Carver College of Medicine, Iowa City, IA, USA
| | - Yuan Lu
- Department of Psychiatry, University of Iowa, Carver College of Medicine, Iowa City, IA, USA
| | - Rachel M Anderson
- Department of Psychology, University of Iowa, Carver College of Medicine, Iowa City, IA, USA
| | - Michael Z Khan
- Department of Psychiatry, University of Iowa, Carver College of Medicine, Iowa City, IA, USA
| | - Varun Nath
- Department of Psychiatry, University of Iowa, Carver College of Medicine, Iowa City, IA, USA
| | - Latisha McDaniel
- Department of Psychiatry, University of Iowa, Carver College of Medicine, Iowa City, IA, USA
| | - Michael Lutter
- Department of Psychiatry, University of Iowa, Carver College of Medicine, Iowa City, IA, USA
| | - Jason J Radley
- Department of Psychology, University of Iowa, Carver College of Medicine, Iowa City, IA, USA
| | - Andrew A Pieper
- Department of Psychiatry, University of Iowa, Carver College of Medicine, Iowa City, IA, USA; Department of Neurology, University of Iowa, Carver College of Medicine, Iowa City, IA, USA; Free Radical & Radiation Biology Program, Department of Radiation Oncology Holden Comprehensive Cancer Center, University of Iowa, Carver College of Medicine, Iowa City, IA, USA
| | - Huxing Cui
- Department of Psychiatry, University of Iowa, Carver College of Medicine, Iowa City, IA, USA.
| |
Collapse
|
139
|
Deng QQ, Sheng WL, Zhang G, Weng SJ, Yang XL, Zhong YM. Signalling mechanism for somatostatin receptor 5-mediated suppression of AMPA responses in rat retinal ganglion cells. Neuropharmacology 2016; 107:215-226. [DOI: 10.1016/j.neuropharm.2016.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 01/25/2016] [Accepted: 03/02/2016] [Indexed: 01/21/2023]
|
140
|
Abstract
Regulation of AMPA receptor (AMPAR) function is a fundamental mechanism controlling synaptic strength during long-term potentiation/depression and homeostatic scaling. AMPAR function and membrane trafficking is controlled by protein-protein interactions, as well as by posttranslational modifications. Phosphorylation of the GluA1 AMPAR subunit at S845 and S831 play especially important roles during synaptic plasticity. Recent controversy has emerged regarding the extent to which GluA1 phosphorylation may contribute to synaptic plasticity. Here we used a variety of methods to measure the population of phosphorylated GluA1-containing AMPARs in cultured primary neurons and mouse forebrain. Phosphorylated GluA1 represents large fractions from 12% to 50% of the total population under basal and stimulated conditions in vitro and in vivo. Furthermore, a large fraction of synapses are positive for phospho-GluA1-containing AMPARs. Our results support the large body of research indicating a prominent role of GluA1 phosphorylation in synaptic plasticity.
Collapse
|
141
|
Li YL, Zhou J, Zhang H, Luo Y, Long LH, Hu ZL, Chen JG, Wang F, Wu PF. Hydrogen Sulfide Promotes Surface Insertion of Hippocampal AMPA Receptor GluR1 Subunit via Phosphorylating at Serine-831/Serine-845 Sites Through a Sulfhydration-Dependent Mechanism. CNS Neurosci Ther 2016; 22:789-98. [PMID: 27380893 DOI: 10.1111/cns.12585] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Revised: 05/13/2016] [Accepted: 06/10/2016] [Indexed: 12/14/2022] Open
Abstract
AIMS Hydrogen sulfide (H2 S) has been widely accepted as a gas neuromodulator to regulate synaptic function. Herein, we set out to determine the effect of H2 S on α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor (AMPAR) and its mechanism. METHODS BS(3) protein cross-linking, Western blot, patch clamp, and biotin-switch assay. RESULTS Bath application of H2 S donor NaHS (50 and 100 μM) rapidly promoted surface insertion of hippocampal AMPAR GluR1 subunit. This effect can be abolished by dithiothreitol (DTT) and mimicked by Na2 S4 , indicating that a sulfhydration-dependent mechanism may be involved. NaHS increased APMAR-mediated EPSC and led to an elevation of GluR2-lacking AMPAR content. Notably, NaHS did not increase the sulfhydration of AMPAR subunits, but it significantly increased the phosphorylation of GluR1 at serine-831 and serine-845 sites. Postsynaptic signal pathways that control GluR1 phosphorylation, such as protein kinase A (PKA), protein kinase C, and calcium/calmodulin-dependent protein kinases II (CaMKII), were sulfhydrated, activated by NaHS, and these effects can be occluded by DTT. H2 S increased S-sulfhydration of protein phosphatase type 2A (PP2A), which may be partially involved in the activation of signal pathways. CONCLUSION Our data suggest that H2 S promotes surface insertion of AMPARs via phosphorylation of GluR1, which depends on a sulfhydration-mediated mechanism.
Collapse
Affiliation(s)
- Yuan-Long Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jun Zhou
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hai Zhang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yi Luo
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Li-Hong Long
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, Hubei, China.,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei, China.,Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhuang-Li Hu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, Hubei, China.,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei, China.,Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jian-Guo Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, Hubei, China.,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei, China.,Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fang Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, Hubei, China.,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei, China.,Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Peng-Fei Wu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, Hubei, China.,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei, China.,Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
142
|
Structural rearrangement of the intracellular domains during AMPA receptor activation. Proc Natl Acad Sci U S A 2016; 113:E3950-9. [PMID: 27313205 DOI: 10.1073/pnas.1601747113] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) are ligand-gated ion channels that mediate the majority of fast excitatory neurotransmission in the central nervous system. Despite recent advances in structural studies of AMPARs, information about the specific conformational changes that underlie receptor function is lacking. Here, we used single and dual insertion of GFP variants at various positions in AMPAR subunits to enable measurements of conformational changes using fluorescence resonance energy transfer (FRET) in live cells. We produced dual CFP/YFP-tagged GluA2 subunit constructs that had normal activity and displayed intrareceptor FRET. We used fluorescence lifetime imaging microscopy (FLIM) in live HEK293 cells to determine distinct steady-state FRET efficiencies in the presence of different ligands, suggesting a dynamic picture of the resting state. Patch-clamp fluorometry of the double- and single-insert constructs showed that both the intracellular C-terminal domain (CTD) and the loop region between the M1 and M2 helices move during activation and the CTD is detached from the membrane. Our time-resolved measurements revealed unexpectedly complex fluorescence changes within these intracellular domains, providing clues as to how posttranslational modifications and receptor function interact.
Collapse
|
143
|
Kibaly C, Kam AY, Loh HH, Law PY. Naltrexone Facilitates Learning and Delays Extinction by Increasing AMPA Receptor Phosphorylation and Membrane Insertion. Biol Psychiatry 2016; 79:906-16. [PMID: 26049209 PMCID: PMC4630208 DOI: 10.1016/j.biopsych.2015.04.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 04/09/2015] [Accepted: 04/11/2015] [Indexed: 01/09/2023]
Abstract
BACKGROUND The opioid antagonists naloxone/naltrexone are involved in improving learning and memory, but their cellular and molecular mechanisms remain unknown. We investigated the effect of naloxone/naltrexone on hippocampal α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) trafficking, a molecular substrate of learning and memory, as a probable mechanism for the antagonists activity. METHODS To measure naloxone/naltrexone-regulated AMPAR trafficking, pHluorin-GluA1 imaging and biochemical analyses were performed on primary hippocampal neurons. To establish the in vivo role of GluA1-Serine 845 (S845) phosphorylation on the behavioral effect induced by inhibition of the endogenous μ-opioid receptor (MOR) by naltrexone, MOR knockout, and GluA1-S845A mutant (in which Ser(845) was mutated to Ala) mice were tested in a water maze after chronic naltrexone administration. Behavioral responses and GluA1 levels in the hippocampal postsynaptic density in wild-type and GluA1-S845A mutant mice were compared using western blot analysis. RESULTS In vitro prolonged naloxone/naltrexone exposure significantly increased synaptic and extrasynaptic GluA1 membrane expression as well as GluA1-S845 phosphorylation. In the MOR knockout and GluA1-S845A mutant mice, naltrexone did not improve learning, which suggests that naltrexone acts via inhibition of endogenous MOR action and alteration of GluA1 phosphorylation. Naltrexone-treated wild-type mice had significantly increased phosphorylated GluA1-S845 and GluA1 levels in their hippocampal postsynaptic density on the third day of acquisition, which is the time when naltrexone significantly improved learning. CONCLUSIONS The beneficial effect of naltrexone on spatial learning and memory under normal conditions appears to be the result of increasing GluA1-S845 phosphorylation-dependent AMPAR trafficking. These results can be further explored in a mouse model of memory loss.
Collapse
Affiliation(s)
- Cherkaouia Kibaly
- Department of Pharmacology and Basic Research Center on Molecular and Cell Biology of Drug Addiction, University of Minnesota, Minneapolis, Minnesota.
| | | | | | | |
Collapse
|
144
|
Abstract
AMPA receptors (AMPARs) are assemblies of four core subunits, GluA1-4, that mediate most fast excitatory neurotransmission. The component subunits determine the functional properties of AMPARs, and the prevailing view is that the subunit composition also determines AMPAR trafficking, which is dynamically regulated during development, synaptic plasticity and in response to neuronal stress in disease. Recently, the subunit dependence of AMPAR trafficking has been questioned, leading to a reappraisal of this field. In this Review, we discuss what is known, uncertain, conjectured and unknown about the roles of the individual subunits, and how they affect AMPAR assembly, trafficking and function under both normal and pathological conditions.
Collapse
|
145
|
Woods CA, Guttman ZR, Huang D, Kolaric RA, Rabinowitsch AI, Jones KT, Cabeza de Vaca S, Sclafani A, Carr KD. Insulin receptor activation in the nucleus accumbens reflects nutritive value of a recently ingested meal. Physiol Behav 2016; 159:52-63. [PMID: 26988281 DOI: 10.1016/j.physbeh.2016.03.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 03/01/2016] [Accepted: 03/12/2016] [Indexed: 12/19/2022]
Abstract
With respect to feeding, insulin is typically thought of as a satiety hormone, acting in the hypothalamus to limit ingestive behavior. However, accumulating evidence suggests that insulin also has the ability to alter dopamine release in the striatum and influence food preferences. With increased access to high calorie foods, Western societies have a high prevalence of obesity, accompanied by insulin insensitivity. Little is known about how insulin is trafficked into the brain following food consumption and whether insulin insensitivity in the periphery is mirrored in the central nervous system. We investigated insulin receptor activation in the ventral striatum of rats receiving water or 16% glucose either orally or intragastrically. We also investigated whether glucose-induced insulin receptor activation was altered in food-restricted (FR) or diet-induced obesity (OB) rat models. Lastly, we examined whether insulin plays a significant role in flavor-nutrient preference learning. Glucose intake stimulated a rapid increase in insulin receptor activity in the ventral striatum of FR and ad libitum (AL) fed rats, but not OB rats. Similarly, both AL and FR, but not OB rats demonstrated significant flavor-nutrient preferences. However AL rats receiving brief inhibition of insulin activity during conditioning failed to acquire a significant flavor-nutrient preference. These findings suggest that impaired insulin receptor activation in the ventral striatum may result in inaccurate valuation of nutritive foods, which could lead to overconsumption of food or the selection of foods that don't accurately meet the body's current physiological needs.
Collapse
Affiliation(s)
- C A Woods
- Center for Neural Science, NYU Graduate School of Arts & Sciences, United States
| | - Z R Guttman
- Center for Neural Science, NYU Graduate School of Arts & Sciences, United States
| | - D Huang
- Dept of Psychiatry, NYU School of Medicine, United States
| | - R A Kolaric
- Dept of Psychiatry, NYU School of Medicine, United States
| | | | - K T Jones
- Dept of Psychiatry, NYU School of Medicine, United States
| | | | - A Sclafani
- Dept of Psychology, Brooklyn College, CUNY, United States
| | - K D Carr
- Center for Neural Science, NYU Graduate School of Arts & Sciences, United States; Dept of Psychiatry, NYU School of Medicine, United States; Dept of Biochemistry and Molecular Pharmacology, NYU School of Medicine, United States
| |
Collapse
|
146
|
Di Maio V, Ventriglia F, Santillo S. A model of dopamine regulation of glutamatergic synapse on medium size spiny neurons. Biosystems 2016; 142-143:25-31. [PMID: 26957078 DOI: 10.1016/j.biosystems.2016.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 02/10/2016] [Accepted: 03/01/2016] [Indexed: 11/24/2022]
Abstract
Spiny neurons of striatum receive glutamatergic synapses on dendritic spines on the neck of which project dopaminergic synapses. Dopamine modulates, by D1 type receptors, the glutamatergic synapses by inducing the phosphorylation of AMPA and NMDA receptors which produces an increased amplitude response. Herein we present a model where, in addition to phosphorylation, the direct modulation by dopamine of the spine resistance can cooperate in producing the observed effect on some of these synapses.
Collapse
Affiliation(s)
- Vito Di Maio
- Istituto di Scienze Applicate e Sistemi Intelligenti del CNR, Italy.
| | | | - Silvia Santillo
- Istituto di Scienze Applicate e Sistemi Intelligenti del CNR, Italy.
| |
Collapse
|
147
|
Jiang L, Voulalas P, Ji Y, Masri R. Post-translational modification of cortical GluA receptors in rodents following spinal cord lesion. Neuroscience 2016; 316:122-9. [PMID: 26724583 PMCID: PMC4724505 DOI: 10.1016/j.neuroscience.2015.12.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 12/17/2015] [Accepted: 12/21/2015] [Indexed: 11/23/2022]
Abstract
Previous studies investigating the pathophysiology of neuropathic pain caused by injury to the spinal cord suggest that pain may result, at least in part, from maladaptive plasticity in the somatosensory cortex and associated pain networks. However, little is known about the molecular and cellular mechanisms leading to maladaptive plasticity in the cortex and how they contribute to the development of neuropathic pain. AMPA-type glutamate receptors (GluARs) mediate fast excitatory synaptic transmission in the mammalian brain and play an important role in pain processing. Here we used an electrolytic lesion model of spinal cord injury in animals to study the expression and phosphorylation of GluA1 and 2 in the primary somatosensory cortex (S1). Experiments in rats and mice revealed that maladaptive plasticity and hypersensitivity after spinal cord lesion (SCL) are associated with a reduction in the fraction of GluA1 subunits that are phosphorylated at serine 831 (S831) in the hindlimb representation of S1 (S1HL). Manipulations that reduce the fraction of phosphorylated S831 in S1HL of non-lesioned animals, including low-frequency electrical stimulation and viral-mediated gene transfer of mutant S831, were associated with the development of hypersensitivity. Taken together, these findings suggest that phosphorylation of GluA1 at S831 plays an important role in the development of hypersensitivity after SCL.
Collapse
Affiliation(s)
- L Jiang
- Department of Endodontics, Periodontics, and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, United States
| | - P Voulalas
- Department of Endodontics, Periodontics, and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, United States
| | - Y Ji
- Department of Endodontics, Periodontics, and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, United States
| | - R Masri
- Department of Endodontics, Periodontics, and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, United States; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, United States.
| |
Collapse
|
148
|
O'Leary H, Bernard PB, Castano AM, Benke TA. Enhanced long term potentiation and decreased AMPA receptor desensitization in the acute period following a single kainate induced early life seizure. Neurobiol Dis 2015; 87:134-44. [PMID: 26706598 DOI: 10.1016/j.nbd.2015.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 11/17/2015] [Accepted: 12/14/2015] [Indexed: 02/04/2023] Open
Abstract
Neonatal seizures are associated with long term disabilities including epilepsy and cognitive deficits. Using a neonatal seizure rat model that does not develop epilepsy, but develops a phenotype consistent with other models of intellectual disability (ID) and autism spectrum disorders (ASD), we sought to isolate the acute effects of a single episode of early life seizure on hippocampal CA1 synaptic development and plasticity. We have previously shown chronic changes in glutamatergic synapses, loss of long term potentiation (LTP) and enhanced long term depression (LTD), in the adult male rat ~50days following kainic acid (KA) induced early life seizure (KA-ELS) in post-natal (P) 7day old male Sprague-Dawley rats. In the present work, we examined the electrophysiological properties and expression levels of glutamate receptors in the acute period, 2 and 7days, post KA-ELS. Our results show for the first time enhanced LTP 7days after KA-ELS, but no change 2days post KA-ELS. Additionally, we report that ionotropic α-amino-3-hydroxy-5-methyl-isoxazole-propionic acid type glutamate receptor (AMPAR) desensitization is decreased in the same time frame, with no changes in AMPAR expression, phosphorylation, or membrane insertion. Inappropriate enhancement of the synaptic connections in the acute period after the seizure could alter the normal patterning of synaptic development in the hippocampus during this critical period and contribute to learning deficits. Thus, this study demonstrates a novel mechanism by which KA-ELS alters early network properties that potentially lead to adverse outcomes.
Collapse
Affiliation(s)
- Heather O'Leary
- Department of Pediatrics, University of Colorado, School of Medicine, 80045, USA
| | - Paul B Bernard
- Department of Pediatrics, University of Colorado, School of Medicine, 80045, USA
| | - Anna M Castano
- Department of Pediatrics, University of Colorado, School of Medicine, 80045, USA
| | - Tim A Benke
- Department of Pediatrics, University of Colorado, School of Medicine, 80045, USA; Department of Neurology, University of Colorado, School of Medicine, 80045, USA; Department of Pharmacology, University of Colorado, School of Medicine, 80045, USA; Department of Otolaryngology, University of Colorado, School of Medicine, 80045, USA; Neuroscience Graduate Program, University of Colorado, School of Medicine, 80045, USA.
| |
Collapse
|
149
|
Ichinose S, Ogawa T, Hirokawa N. Mechanism of Activity-Dependent Cargo Loading via the Phosphorylation of KIF3A by PKA and CaMKIIa. Neuron 2015; 87:1022-35. [PMID: 26335646 DOI: 10.1016/j.neuron.2015.08.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 06/30/2015] [Accepted: 07/31/2015] [Indexed: 12/14/2022]
Abstract
A regulated mechanism of cargo loading is crucial for intracellular transport. N-cadherin, a synaptic adhesion molecule that is critical for neuronal function, must be precisely transported to dendritic spines in response to synaptic activity and plasticity. However, the mechanism of activity-dependent cargo loading remains unclear. To elucidate this mechanism, we investigated the activity-dependent transport of N-cadherin via its transporter, KIF3A. First, by comparing KIF3A-bound cargo vesicles with unbound KIF3A, we identified critical KIF3A phosphorylation sites and specific kinases, PKA and CaMKIIa, using quantitative phosphoanalyses. Next, mutagenesis and kinase inhibitor experiments revealed that N-cadherin transport was enhanced via phosphorylation of the KIF3A C terminus, thereby increasing cargo-loading activity. Furthermore, N-cadherin transport was enhanced during homeostatic upregulation of synaptic strength, triggered by chronic inactivation by TTX. We propose the first model of activity-dependent cargo loading, in which phosphorylation of the KIF3A C terminus upregulates the loading and transport of N-cadherin in homeostatic synaptic plasticity.
Collapse
Affiliation(s)
- Sotaro Ichinose
- Department of Cell Biology and Anatomy, Graduate School of Medicine, University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tadayuki Ogawa
- Department of Cell Biology and Anatomy, Graduate School of Medicine, University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Nobutaka Hirokawa
- Department of Cell Biology and Anatomy, Graduate School of Medicine, University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan; Center of Excellence in Genome Medicine Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
150
|
Di Angelantonio S, Bertollini C, Piccinin S, Rosito M, Trettel F, Pagani F, Limatola C, Ragozzino D. Basal adenosine modulates the functional properties of AMPA receptors in mouse hippocampal neurons through the activation of A1R A2AR and A3R. Front Cell Neurosci 2015; 9:409. [PMID: 26528137 PMCID: PMC4601258 DOI: 10.3389/fncel.2015.00409] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 09/25/2015] [Indexed: 11/30/2022] Open
Abstract
Adenosine is a widespread neuromodulator within the CNS and its extracellular level is increased during hypoxia or intense synaptic activity, modulating pre- and postsynaptic sites. We studied the neuromodulatory action of adenosine on glutamatergic currents in the hippocampus, showing that activation of multiple adenosine receptors (ARs) by basal adenosine impacts postsynaptic site. Specifically, the stimulation of both A1R and A3R reduces AMPA currents, while A2AR has an opposite potentiating effect. The effect of ARs stimulation on glutamatergic currents in hippocampal cultures was investigated using pharmacological and genetic approaches. A3R inhibition by MRS1523 increased GluR1-Ser845 phosphorylation and potentiated AMPA current amplitude, increasing the apparent affinity for the agonist. A similar effect was observed blocking A1R with DPCPX or by genetic deletion of either A3R or A1R. Conversely, impairment of A2AR reduced AMPA currents, and decreased agonist sensitivity. Consistently, in hippocampal slices, ARs activation by AR agonist NECA modulated glutamatergic current amplitude evoked by AMPA application or afferent fiber stimulation. Opposite effects of AR subtypes stimulation are likely associated to changes in GluR1 phosphorylation and represent a novel mechanism of physiological modulation of glutamatergic transmission by adenosine, likely acting in normal conditions in the brain, depending on the level of extracellular adenosine and the distribution of AR subtypes.
Collapse
Affiliation(s)
- Silvia Di Angelantonio
- Istituto Pasteur-Fondazione Cenci Bolognetti and Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma Roma, Italy ; Center for Life Nanoscience, Istituto Italiano di Tecnologia Rome, Italy
| | - Cristina Bertollini
- Istituto Pasteur-Fondazione Cenci Bolognetti and Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma Roma, Italy
| | - Sonia Piccinin
- Istituto Pasteur-Fondazione Cenci Bolognetti and Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma Roma, Italy
| | - Maria Rosito
- Istituto Pasteur-Fondazione Cenci Bolognetti and Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma Roma, Italy
| | - Flavia Trettel
- Istituto Pasteur-Fondazione Cenci Bolognetti and Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma Roma, Italy
| | - Francesca Pagani
- Center for Life Nanoscience, Istituto Italiano di Tecnologia Rome, Italy
| | - Cristina Limatola
- Istituto Pasteur-Fondazione Cenci Bolognetti and Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma Roma, Italy ; Neuromed, Istituto di Ricovero e Cura a Carattere Scientifico Pozzilli, Italy
| | - Davide Ragozzino
- Istituto Pasteur-Fondazione Cenci Bolognetti and Dipartimento di Fisiologia e Farmacologia, Sapienza Università di Roma Roma, Italy ; Neuromed, Istituto di Ricovero e Cura a Carattere Scientifico Pozzilli, Italy
| |
Collapse
|