1
|
Nicoll RA, Schulman H. Synaptic memory and CaMKII. Physiol Rev 2023; 103:2877-2925. [PMID: 37290118 PMCID: PMC10642921 DOI: 10.1152/physrev.00034.2022] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 04/26/2023] [Accepted: 04/30/2023] [Indexed: 06/10/2023] Open
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) and long-term potentiation (LTP) were discovered within a decade of each other and have been inextricably intertwined ever since. However, like many marriages, it has had its up and downs. Based on the unique biochemical properties of CaMKII, it was proposed as a memory molecule before any physiological linkage was made to LTP. However, as reviewed here, the convincing linkage of CaMKII to synaptic physiology and behavior took many decades. New technologies were critical in this journey, including in vitro brain slices, mouse genetics, single-cell molecular genetics, pharmacological reagents, protein structure, and two-photon microscopy, as were new investigators attracted by the exciting challenge. This review tracks this journey and assesses the state of this marriage 40 years on. The collective literature impels us to propose a relatively simple model for synaptic memory involving the following steps that drive the process: 1) Ca2+ entry through N-methyl-d-aspartate (NMDA) receptors activates CaMKII. 2) CaMKII undergoes autophosphorylation resulting in constitutive, Ca2+-independent activity and exposure of a binding site for the NMDA receptor subunit GluN2B. 3) Active CaMKII translocates to the postsynaptic density (PSD) and binds to the cytoplasmic C-tail of GluN2B. 4) The CaMKII-GluN2B complex initiates a structural rearrangement of the PSD that may involve liquid-liquid phase separation. 5) This rearrangement involves the PSD-95 scaffolding protein, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs), and their transmembrane AMPAR-regulatory protein (TARP) auxiliary subunits, resulting in an accumulation of AMPARs in the PSD that underlies synaptic potentiation. 6) The stability of the modified PSD is maintained by the stability of the CaMKII-GluN2B complex. 7) By a process of subunit exchange or interholoenzyme phosphorylation CaMKII maintains synaptic potentiation in the face of CaMKII protein turnover. There are many other important proteins that participate in enlargement of the synaptic spine or modulation of the steps that drive and maintain the potentiation. In this review we critically discuss the data underlying each of the steps. As will become clear, some of these steps are more firmly grounded than others, and we provide suggestions as to how the evidence supporting these steps can be strengthened or, based on the new data, be replaced. Although the journey has been a long one, the prospect of having a detailed cellular and molecular understanding of learning and memory is at hand.
Collapse
Affiliation(s)
- Roger A Nicoll
- Department of Cellular and Molecular Pharmacology, University of California at San Francisco, San Francisco, California, United States
| | - Howard Schulman
- Department of Neurobiology, Stanford University School of Medicine, Stanford, California, United States
- Panorama Research Institute, Sunnyvale, California, United States
| |
Collapse
|
2
|
Perkins EM, Burr K, Banerjee P, Mehta AR, Dando O, Selvaraj BT, Suminaite D, Nanda J, Henstridge CM, Gillingwater TH, Hardingham GE, Wyllie DJA, Chandran S, Livesey MR. Altered network properties in C9ORF72 repeat expansion cortical neurons are due to synaptic dysfunction. Mol Neurodegener 2021; 16:13. [PMID: 33663561 PMCID: PMC7931347 DOI: 10.1186/s13024-021-00433-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 02/14/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Physiological disturbances in cortical network excitability and plasticity are established and widespread in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) patients, including those harbouring the C9ORF72 repeat expansion (C9ORF72RE) mutation - the most common genetic impairment causal to ALS and FTD. Noting that perturbations in cortical function are evidenced pre-symptomatically, and that the cortex is associated with widespread pathology, cortical dysfunction is thought to be an early driver of neurodegenerative disease progression. However, our understanding of how altered network function manifests at the cellular and molecular level is not clear. METHODS To address this we have generated cortical neurons from patient-derived iPSCs harbouring C9ORF72RE mutations, as well as from their isogenic expansion-corrected controls. We have established a model of network activity in these neurons using multi-electrode array electrophysiology. We have then mechanistically examined the physiological processes underpinning network dysfunction using a combination of patch-clamp electrophysiology, immunocytochemistry, pharmacology and transcriptomic profiling. RESULTS We find that C9ORF72RE causes elevated network burst activity, associated with enhanced synaptic input, yet lower burst duration, attributable to impaired pre-synaptic vesicle dynamics. We also show that the C9ORF72RE is associated with impaired synaptic plasticity. Moreover, RNA-seq analysis revealed dysregulated molecular pathways impacting on synaptic function. All molecular, cellular and network deficits are rescued by CRISPR/Cas9 correction of C9ORF72RE. Our study provides a mechanistic view of the early dysregulated processes that underpin cortical network dysfunction in ALS-FTD. CONCLUSION These findings suggest synaptic pathophysiology is widespread in ALS-FTD and has an early and fundamental role in driving altered network function that is thought to contribute to neurodegenerative processes in these patients. The overall importance is the identification of previously unidentified defects in pre and postsynaptic compartments affecting synaptic plasticity, synaptic vesicle stores, and network propagation, which directly impact upon cortical function.
Collapse
Affiliation(s)
- Emma M. Perkins
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, EH16 4SB UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Karen Burr
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, EH16 4SB UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, EH16 4SB UK
| | - Poulomi Banerjee
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, EH16 4SB UK
| | - Arpan R. Mehta
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, EH16 4SB UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, EH16 4SB UK
| | - Owen Dando
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, EH16 4SB UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Bhuvaneish T. Selvaraj
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, EH16 4SB UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, EH16 4SB UK
| | - Daumante Suminaite
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Jyoti Nanda
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, EH16 4SB UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, EH16 4SB UK
| | - Christopher M. Henstridge
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, EH16 4SB UK
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, DD1 9SY UK
| | - Thomas H. Gillingwater
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, EH16 4SB UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Giles E. Hardingham
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, EH16 4SB UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - David J. A. Wyllie
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK
- Centre for Brain Development and Repair, inStem, Bangalore, 560065 India
| | - Siddharthan Chandran
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, EH16 4SB UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, EH16 4SB UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK
- Centre for Brain Development and Repair, inStem, Bangalore, 560065 India
| | - Matthew R. Livesey
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, EH16 4SB UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, S10 2HQ UK
| |
Collapse
|
3
|
Purkey AM, Dell’Acqua ML. Phosphorylation-Dependent Regulation of Ca 2+-Permeable AMPA Receptors During Hippocampal Synaptic Plasticity. Front Synaptic Neurosci 2020; 12:8. [PMID: 32292336 PMCID: PMC7119613 DOI: 10.3389/fnsyn.2020.00008] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/18/2020] [Indexed: 01/28/2023] Open
Abstract
Experience-dependent learning and memory require multiple forms of plasticity at hippocampal and cortical synapses that are regulated by N-methyl-D-aspartate receptors (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type ionotropic glutamate receptors (NMDAR, AMPAR). These plasticity mechanisms include long-term potentiation (LTP) and depression (LTD), which are Hebbian input-specific mechanisms that rapidly increase or decrease AMPAR synaptic strength at specific inputs, and homeostatic plasticity that globally scales-up or -down AMPAR synaptic strength across many or even all inputs. Frequently, these changes in synaptic strength are also accompanied by a change in the subunit composition of AMPARs at the synapse due to the trafficking to and from the synapse of receptors lacking GluA2 subunits. These GluA2-lacking receptors are most often GluA1 homomeric receptors that exhibit higher single-channel conductance and are Ca2+-permeable (CP-AMPAR). This review article will focus on the role of protein phosphorylation in regulation of GluA1 CP-AMPAR recruitment and removal from hippocampal synapses during synaptic plasticity with an emphasis on the crucial role of local signaling by the cAMP-dependent protein kinase (PKA) and the Ca2+calmodulin-dependent protein phosphatase 2B/calcineurin (CaN) that is coordinated by the postsynaptic scaffold protein A-kinase anchoring protein 79/150 (AKAP79/150).
Collapse
Affiliation(s)
| | - Mark L. Dell’Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
4
|
Kloc ML, Pradier B, Chirila AM, Kauer JA. NMDA receptor activation induces long-term potentiation of glycine synapses. PLoS One 2019; 14:e0222066. [PMID: 31498817 PMCID: PMC6733442 DOI: 10.1371/journal.pone.0222066] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 08/21/2019] [Indexed: 01/12/2023] Open
Abstract
Of the fast ionotropic synapses, glycinergic synapses are the least well understood, but are vital for the maintenance of inhibitory signaling in the brain and spinal cord. Glycinergic signaling comprises half of the inhibitory signaling in the spinal cord, and glycinergic synapses are likely to regulate local nociceptive processing as well as the transmission to the brain of peripheral nociceptive information. Here we have investigated the rapid and prolonged potentiation of glycinergic synapses in the superficial dorsal horn of young male and female mice after brief activation of NMDA receptors (NMDARs). Glycinergic inhibitory postsynaptic currents (IPSCs) evoked with lamina II-III stimulation in identified GABAergic neurons in lamina II were potentiated by bath-applied Zn2+ and were depressed by the prostaglandin PGE2, consistent with the presence of both GlyRα1- and GlyRα3-containing receptors. NMDA application rapidly potentiated synaptic glycinergic currents. Whole-cell currents evoked by exogenous glycine were also readily potentiated by NMDA, indicating that the potentiation results from altered numbers or conductance of postsynaptic glycine receptors. Repetitive depolarization alone of the postsynaptic GABAergic neuron also potentiated glycinergic synapses, and intracellular EGTA prevented both NMDA-induced and depolarization-induced potentiation of glycinergic IPSCs. Optogenetic activation of trpv1 lineage afferents also triggered NMDAR-dependent potentiation of glycinergic synapses. Our results suggest that during peripheral injury or inflammation, nociceptor firing during injury is likely to potentiate glycinergic synapses on GABAergic neurons. This disinhibition mechanism may be engaged rapidly, altering dorsal horn circuitry to promote the transmission of nociceptive information to the brain.
Collapse
Affiliation(s)
- Michelle L. Kloc
- Dept. of Pharmacology, Physiology and Biotechnology, Brown Institute for Brain Science, Brown University, Providence, RI, United States of America
| | - Bruno Pradier
- Dept. of Pharmacology, Physiology and Biotechnology, Brown Institute for Brain Science, Brown University, Providence, RI, United States of America
| | - Anda M. Chirila
- Dept. of Pharmacology, Physiology and Biotechnology, Brown Institute for Brain Science, Brown University, Providence, RI, United States of America
| | - Julie A. Kauer
- Dept. of Pharmacology, Physiology and Biotechnology, Brown Institute for Brain Science, Brown University, Providence, RI, United States of America
- * E-mail: ,
| |
Collapse
|
5
|
Deletion of LRRTM1 and LRRTM2 in adult mice impairs basal AMPA receptor transmission and LTP in hippocampal CA1 pyramidal neurons. Proc Natl Acad Sci U S A 2018; 115:E5382-E5389. [PMID: 29784826 DOI: 10.1073/pnas.1803280115] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Leucine-rich repeat transmembrane (LRRTM) proteins are synaptic cell adhesion molecules that influence synapse formation and function. They are genetically associated with neuropsychiatric disorders, and via their synaptic actions likely regulate the establishment and function of neural circuits in the mammalian brain. Here, we take advantage of the generation of a LRRTM1 and LRRTM2 double conditional knockout mouse (LRRTM1,2 cKO) to examine the role of LRRTM1,2 at mature excitatory synapses in hippocampal CA1 pyramidal neurons. Genetic deletion of LRRTM1,2 in vivo in CA1 neurons using Cre recombinase-expressing lentiviruses dramatically impaired long-term potentiation (LTP), an impairment that was rescued by simultaneous expression of LRRTM2, but not LRRTM4. Mutation or deletion of the intracellular tail of LRRTM2 did not affect its ability to rescue LTP, while point mutations designed to impair its binding to presynaptic neurexins prevented rescue of LTP. In contrast to previous work using shRNA-mediated knockdown of LRRTM1,2, KO of these proteins at mature synapses also caused a decrease in AMPA receptor-mediated, but not NMDA receptor-mediated, synaptic transmission and had no detectable effect on presynaptic function. Imaging of recombinant photoactivatable AMPA receptor subunit GluA1 in the dendritic spines of cultured neurons revealed that it was less stable in the absence of LRRTM1,2. These results illustrate the advantages of conditional genetic deletion experiments for elucidating the function of endogenous synaptic proteins and suggest that LRRTM1,2 proteins help stabilize synaptic AMPA receptors at mature spines during basal synaptic transmission and LTP.
Collapse
|
6
|
Interplay between global and pathway-specific synaptic plasticity in CA1 pyramidal cells. Sci Rep 2017; 7:17040. [PMID: 29213058 PMCID: PMC5719010 DOI: 10.1038/s41598-017-17161-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 11/22/2017] [Indexed: 12/22/2022] Open
Abstract
Mechanisms underlying information storage have been depicted for global cell-wide and pathway-specific synaptic plasticity. Yet, little is known how these forms of plasticity interact to enhance synaptic competition and network stability. We examined synaptic interactions between apical and basal dendrites of CA1 pyramidal neurons in mouse hippocampal slices. Bursts (50 Hz) of three action potentials (AP-bursts) paired with preceding presynaptic stimulation in stratum radiatum specifically led to LTP of the paired pathway in adult mice (P75). At adolescence (P28), an increase in burst frequency (>50 Hz) was required to gain timing-dependent LTP. Surprisingly, paired radiatum and unpaired oriens pathway potentiated, unless the pre-post delay was shortened from 10 to 5 ms, which selectively potentiated paired radiatum pathway, since unpaired oriens pathway decreased back to baseline. Conversely, the exact same 5 ms pairing in stratum oriens potentiated both pathways, as did AP-bursts alone, which potentiated synaptic efficacy as well as current-evoked postsynaptic spiking. L-type voltage-gated Ca2+ channels were involved in mediating synaptic potentiation in oriens, whereas NMDA and adenosine receptors counteracted unpaired stratum oriens potentiation following pairing in stratum radiatum. This asymmetric plasticity uncovers important insights into alterations of synaptic efficacy and intrinsic neuronal excitability for pathways that convey hippocampal and extra-hippocampal information.
Collapse
|
7
|
The Retromer Supports AMPA Receptor Trafficking During LTP. Neuron 2017; 94:74-82.e5. [PMID: 28384478 DOI: 10.1016/j.neuron.2017.03.020] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 01/24/2017] [Accepted: 03/10/2017] [Indexed: 02/07/2023]
Abstract
Alterations in the function of the retromer, a multisubunit protein complex that plays a specialized role in endosomal sorting, have been linked to Alzheimer's and Parkinson's diseases, yet little is known about the retromer's role in the mature brain. Using in vivo knockdown of the critical retromer component VPS35, we demonstrate a specific role for this endosomal sorting complex in the trafficking of AMPA receptors during NMDA-receptor-dependent LTP at mature hippocampal synapses. The impairment of LTP due to VPS35 knockdown was mechanistically independent of any role of the retromer in the production of Aβ from APP. Finally, we find surprising differences between Alzheimer's- and Parkinson's-disease-linked VPS35 mutations in supporting this pathway. These findings demonstrate a key role for the retromer in LTP and provide insights into how retromer malfunction in the mature brain may contribute to symptoms of common neurodegenerative diseases. VIDEO ABSTRACT.
Collapse
|
8
|
Conditional ablation of neuroligin-1 in CA1 pyramidal neurons blocks LTP by a cell-autonomous NMDA receptor-independent mechanism. Mol Psychiatry 2017; 22:375-383. [PMID: 27217145 PMCID: PMC5122464 DOI: 10.1038/mp.2016.80] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 02/26/2016] [Accepted: 03/02/2016] [Indexed: 02/05/2023]
Abstract
Neuroligins are postsynaptic cell-adhesion molecules implicated in autism and other neuropsychiatric disorders. Despite extensive work, the role of neuroligins in synapse function and plasticity, especially N-methyl-d-aspartate (NMDA) receptor (NMDAR)-dependent long-term potentiation (LTP), remains unclear. To establish which synaptic functions unequivocally require neuroligins, we analyzed single and triple conditional knockout (cKO) mice for all three major neuroligin isoforms (NL1-NL3). We inactivated neuroligins by stereotactic viral expression of Cre-recombinase in hippocampal CA1 region pyramidal neurons at postnatal day 0 (P0) or day 21 (P21) and measured synaptic function, synaptic plasticity and spine numbers in acute hippocampal slices 2-3 weeks later. Surprisingly, we find that ablation of neuroligins in newborn or juvenile mice only modestly impaired basal synaptic function in hippocampus and caused no alteration in postsynaptic spine numbers. However, triple cKO of NL1-NL3 or single cKO of NL1 impaired NMDAR-mediated excitatory postsynaptic currents and abolished NMDAR-dependent LTP. Strikingly, the NL1 cKO also abolished LTP elicited by activation of L-type Ca2+-channels during blockade of NMDARs. These findings demonstrate that neuroligins are generally not essential for synapse formation in CA1 pyramidal neurons but shape synaptic properties and that NL1 specifically is required for LTP induced by postsynaptic Ca2+-elevations, a function which may contribute to the pathophysiological role of neuroligins in brain disorders.
Collapse
|
9
|
Lussier MP, Sanz-Clemente A, Roche KW. Dynamic Regulation of N-Methyl-d-aspartate (NMDA) and α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid (AMPA) Receptors by Posttranslational Modifications. J Biol Chem 2015; 290:28596-603. [PMID: 26453298 DOI: 10.1074/jbc.r115.652750] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Many molecular mechanisms underlie the changes in synaptic glutamate receptor content that are required by neuronal networks to generate cellular correlates of learning and memory. During the last decade, posttranslational modifications have emerged as critical regulators of synaptic transmission and plasticity. Notably, phosphorylation, ubiquitination, and palmitoylation control the stability, trafficking, and synaptic expression of glutamate receptors in the central nervous system. In the current review, we will summarize some of the progress made by the neuroscience community regarding our understanding of phosphorylation, ubiquitination, and palmitoylation of the NMDA and AMPA subtypes of glutamate receptors.
Collapse
Affiliation(s)
- Marc P Lussier
- From the Département de Chimie, Université du Québec à Montréal, Montréal, Québec H3C 3P8, Canada
| | - Antonio Sanz-Clemente
- the Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, and
| | - Katherine W Roche
- the Receptor Biology Section, NINDS, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
10
|
Ménard C, Gaudreau P, Quirion R. Signaling pathways relevant to cognition-enhancing drug targets. Handb Exp Pharmacol 2015; 228:59-98. [PMID: 25977080 DOI: 10.1007/978-3-319-16522-6_3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Aging is generally associated with a certain cognitive decline. However, individual differences exist. While age-related memory deficits can be observed in humans and rodents in the absence of pathological conditions, some individuals maintain intact cognitive functions up to an advanced age. The mechanisms underlying learning and memory processes involve the recruitment of multiple signaling pathways and gene expression, leading to adaptative neuronal plasticity and long-lasting changes in brain circuitry. This chapter summarizes the current understanding of how these signaling cascades could be modulated by cognition-enhancing agents favoring memory formation and successful aging. It focuses on data obtained in rodents, particularly in the rat as it is the most common animal model studied in this field. First, we will discuss the role of the excitatory neurotransmitter glutamate and its receptors, downstream signaling effectors [e.g., calcium/calmodulin-dependent protein kinase II (CaMKII), protein kinase C (PKC), extracellular signal-regulated kinases (ERK), mammalian target of rapamycin (mTOR), cAMP response element-binding protein (CREB)], associated immediate early gene (e.g., Homer 1a, Arc and Zif268), and growth factors [insulin-like growth factors (IGFs) and brain-derived neurotrophic factor (BDNF)] in synaptic plasticity and memory formation. Second, the impact of the cholinergic system and related modulators on memory will be briefly reviewed. Finally, since dynorphin neuropeptides have recently been associated with memory impairments in aging, it is proposed as an attractive target to develop novel cognition-enhancing agents.
Collapse
Affiliation(s)
- Caroline Ménard
- Douglas Mental Health University Institute, McGill University, Perry Pavilion, 6875 LaSalle Boulevard, Montreal, QC, Canada, H4H 1R3
| | | | | |
Collapse
|
11
|
Abstract
The study of synaptic plasticity and specifically LTP and LTD is one of the most active areas of research in neuroscience. In the last 25 years we have come a long way in our understanding of the mechanisms underlying synaptic plasticity. In 1988, AMPA and NMDA receptors were not even molecularly identified and we only had a simple model of the minimal requirements for the induction of plasticity. It is now clear that the modulation of the AMPA receptor function and membrane trafficking is critical for many forms of synaptic plasticity and a large number of proteins have been identified that regulate this complex process. Here we review the progress over the last two and a half decades and discuss the future challenges in the field.
Collapse
|
12
|
Ovarian hormone loss impairs excitatory synaptic transmission at hippocampal CA3-CA1 synapses. J Neurosci 2013; 33:16158-69. [PMID: 24107948 DOI: 10.1523/jneurosci.2001-13.2013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Premature and long-term ovarian hormone loss following ovariectomy (OVX) is associated with cognitive impairment. This condition is prevented by estradiol (E2) therapy when initiated shortly following OVX but not after substantial delay. To determine whether these clinical findings are correlated with changes in synaptic functions, we used adult OVX rats to evaluate the consequences of short-term (7-10 d, OVXControl) and long-term (∼5 months, OVXLT) ovarian hormone loss, as well as subsequent in vivo E2 treatment, on excitatory synaptic transmission at the hippocampal CA3-CA1 synapses important for learning and memory. The results show that ovarian hormone loss was associated with a marked decrease in synaptic strength. E2 treatment increased synaptic strength in OVXControl but not OVXLT rats, demonstrating a change in the efficacy for E2 5 months following OVX. E2 also had a more rapid effect: within minutes of bath application, E2 acutely increased synaptic strength in all groups except OVXLT rats that did not receive in vivo E2 treatment. E2's acute effect was mediated postsynaptically, and required Ca(2+) influx through the voltage-gated Ca(2+) channels. Despite E2's acute effect, synaptic strength of OVXLT rats remained significantly lower than that of OVXControl rats. Thus, changes in CA3-CA1 synaptic transmission associated with ovarian hormone loss cannot be fully reversed with delayed E2 treatment. Given that synaptic strength at CA3-CA1 synapses is related to the ability to learn hippocampus-dependent tasks, these findings provide additional insights for understanding cognitive impairment-associated long-term ovarian hormone loss and ineffectiveness for delayed E2 treatment to maintain cognitive functions.
Collapse
|
13
|
Oku S, Takahashi N, Fukata Y, Fukata M. In silico screening for palmitoyl substrates reveals a role for DHHC1/3/10 (zDHHC1/3/11)-mediated neurochondrin palmitoylation in its targeting to Rab5-positive endosomes. J Biol Chem 2013; 288:19816-29. [PMID: 23687301 DOI: 10.1074/jbc.m112.431676] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Protein palmitoylation, a common post-translational lipid modification, plays an important role in protein trafficking and functions. Recently developed palmitoyl-proteomic methods identified many novel substrates. However, the whole picture of palmitoyl substrates has not been clarified. Here, we performed global in silico screening using the CSS-Palm 2.0 program, free software for prediction of palmitoylation sites, and selected 17 candidates as novel palmitoyl substrates. Of the 17 candidates, 10 proteins, including 6 synaptic proteins (Syd-1, transmembrane AMPA receptor regulatory protein (TARP) γ-2, TARP γ-8, cornichon-2, Ca(2+)/calmodulin-dependent protein kinase IIα, and neurochondrin (Ncdn)/norbin), one focal adhesion protein (zyxin), two ion channels (TRPM8 and TRPC1), and one G-protein-coupled receptor (orexin 2 receptor), were palmitoylated. Using the DHHC palmitoylating enzyme library, we found that all tested substrates were palmitoylated by the Golgi-localized DHHC3/7 subfamily. Ncdn, a regulator for neurite outgrowth and synaptic plasticity, was robustly palmitoylated by the DHHC1/10 (zDHHC1/11; z1/11) subfamily, whose substrate has not yet been reported. As predicted by CSS-Palm 2.0, Cys-3 and Cys-4 are the palmitoylation sites for Ncdn. Ncdn was specifically localized in somato-dendritic regions, not in the axon of rat cultured neurons. Stimulated emission depletion microscopy revealed that Ncdn was localized to Rab5-positive early endosomes in a palmitoylation-dependent manner, where DHHC1/10 (z1/11) were also distributed. Knockdown of DHHC1, -3, or -10 (z11) resulted in the loss of Ncdn from Rab5-positive endosomes. Thus, through in silico screening, we demonstrate that Ncdn and the DHHC1/10 (z1/11) and DHHC3/7 subfamilies are novel palmitoyl substrate-enzyme pairs and that Ncdn palmitoylation plays an essential role in its specific endosomal targeting.
Collapse
Affiliation(s)
- Shinichiro Oku
- Division of Membrane Physiology, Department of Cell Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | | | | | | |
Collapse
|
14
|
Glutamate receptor 1 phosphorylation at serine 831 and 845 modulates seizure susceptibility and hippocampal hyperexcitability after early life seizures. J Neurosci 2013; 32:17800-12. [PMID: 23223299 DOI: 10.1523/jneurosci.6121-11.2012] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Neonatal seizures can lead to later life epilepsy and neurobehavioral deficits, and there are no treatments to prevent these sequelae. We showed previously that hypoxia-induced seizures in a neonatal rat model induce rapid phosphorylation of serine-831 (S831) and Serine 845 (S845) sites of the AMPA receptor GluR1 subunit and later neuronal hyperexcitability and epilepsy, suggesting that seizure-induced posttranslational modifications may represent a novel therapeutic target. To unambiguously assess the contribution of these sites, we examined seizure susceptibility in wild-type mice versus transgenic knock-in mice with deficits in GluR1 S831 and S845 phosphorylation [GluR1 double-phosphomutant (GluR1 DPM) mice]. Phosphorylation of the GluR1 S831 and S845 sites was significantly increased in the hippocampus and cortex after a single episode of pentyleneterazol-induced seizures in postnatal day 7 (P7) wild-type mouse pups and that transgenic knock-in mice have a higher threshold and longer latencies to seizures. Like the rat, hypoxic seizures in P9 C57BL/6N wild-type mice resulted in transient increases in GluR1 S831 and GluR1 S845 phosphorylation in cortex and were associated with enhanced seizure susceptibility to later-life kainic-acid-induced seizures. In contrast, later-life seizure susceptibility after hypoxia-induced seizures was attenuated in GluR1 DPM mice, supporting a role for posttranslational modifications in seizure-induced network excitability. Finally, human hippocampal samples from neonatal seizure autopsy cases also showed an increase in GluR1 S831 and S845, supporting the validation of this potential therapeutic target in human tissue.
Collapse
|
15
|
Nuritova F, Frenguelli BG. Putative depolarisation-induced retrograde signalling accelerates the repeated hypoxic depression of excitatory synaptic transmission in area CA1 of rat hippocampus via group I metabotropic glutamate receptors. Neuroscience 2012; 222:159-72. [PMID: 22842516 DOI: 10.1016/j.neuroscience.2012.07.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Revised: 07/16/2012] [Accepted: 07/17/2012] [Indexed: 02/08/2023]
Abstract
Excitatory synaptic transmission in area CA1 of the mammalian hippocampus is rapidly depressed during hypoxia. The depression is largely attributable to an increase in extracellular adenosine and activation of inhibitory adenosine A(1) receptors on presynaptic glutamatergic terminals. However, sequential exposure to hypoxia results in a slower subsequent hypoxic depression of excitatory synaptic transmission, a phenomenon we have previously ascribed to a reduction in the release of extracellular adenosine. In the present study we show that this delayed depression of excitatory postsynaptic currents (EPSCs) to repeated hypoxia can be reversed by a period of postsynaptic depolarisation delivered to an individual CA1 neuron, under whole-cell voltage clamp, between two periods of hypoxia. The depolarisation-induced acceleration of the hypoxic depression of the EPSC is dependent upon postsynaptic Ca(2+) influx, the activation of PKC and is blocked by intracellular application of GDP-β-S and N-ethylmaleimide (NEM), inhibitors of membrane fusion events. In addition, the acceleration of the hypoxic depression of the EPSC was prevented by the GI mGluR antagonist AIDA, but not by the CB1 cannabinoid receptor antagonist AM251. Our results suggest a process initiated in the postsynaptic cell that can influence glutamate release during subsequent metabolic stress. This may reflect a novel neuroprotective strategy potentially involving retrograde release of adenosine and/or glutamate.
Collapse
Affiliation(s)
- F Nuritova
- Neurosciences Institute, Division of Pathology & Neuroscience, University of Dundee, Ninewells Hospital & Medical School, Dundee DD1 9SY, UK
| | | |
Collapse
|
16
|
Abstract
Modern theories on memory storage have mainly focused on Hebbian long-term potentiation (LTP), which requires coincident activation of presynaptic and postsynaptic neurons for its induction. In addition to Hebbian LTP, the roles of non-Hebbian plasticity have also been predicted by some neuronal network models. However, still only a few pieces of evidence have been presented for the presence of such plasticity. In this study, we show in mouse hippocampal slices that LTP can be induced by postsynaptic repetitive depolarization alone in the absence of presynaptic inputs. The induction was dependent on voltage-dependent calcium channels instead of NMDA receptors (NMDARs), whereas the expression mechanism was shared with conventional NMDAR-dependent LTP. During the potentiation, the amplitude of spontaneous EPSCs was increased, suggesting a novel neuron-wide nature of this form of LTP. Furthermore, we also successfully induced LTP with trains of action potentials, which supported the possible existence of depolarizing pulse-induced LTP in vivo. Based on these findings, we suggest a model in which neuron-wide LTP works in concert with synapse-specific Hebbian plasticity to help information processing in memory formation.
Collapse
|
17
|
Early alterations of AMPA receptors mediate synaptic potentiation induced by neonatal seizures. J Neurosci 2008; 28:7979-90. [PMID: 18685023 DOI: 10.1523/jneurosci.1734-08.2008] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The highest incidence of seizures during lifetime is found in the neonatal period and neonatal seizures lead to a propensity for epilepsy and long-term cognitive deficits. Here, we identify potential mechanisms that elucidate a critical role for AMPA receptors (AMPARs) in epileptogenesis during this critical period in the developing brain. In a rodent model of neonatal seizures, we have shown previously that administration of antagonists of the AMPARs during the 48 h after seizures prevents long-term increases in seizure susceptibility and seizure-induced neuronal injury. Hypoxia-induced seizures in postnatal day 10 rats induce rapid and reversible alterations in AMPAR signaling resembling changes implicated previously in models of synaptic potentiation in vitro. Hippocampal slices removed after hypoxic seizures exhibited potentiation of AMPAR-mediated synaptic currents, including an increase in the amplitude and frequency of spontaneous and miniature EPSCs as well as increased synaptic potency. This increased excitability was temporally associated with a rapid increase in phosphorylation at GluR1 S845/S831 and GluR2 S880 sites and increased activity of the protein kinases CaMKII (calcium/calmodulin-dependent protein kinase II), PKA, and PKC, which mediate the phosphorylation of these AMPAR subunits. Postseizure administration of AMPAR antagonists NBQX (2,3-dihydroxy-6-nitro-7-sulfonyl-benzo[f]quinoxaline), topiramate, or GYKI-53773 [(1)-1-(4-aminophenyl)-3-acetyl-4-methyl-7,8-methylenedioxy-3,4-dihydro-5H-2,3-benzodiazepine] attenuated the AMPAR potentiation, phosphorylation, and kinase activation and prevented the concurrent increase in in vivo seizure susceptibility. Thus, the potentiation of AMPAR-containing synapses is a reversible, early step in epileptogenesis that offers a novel therapeutic target in the highly seizure-prone developing brain.
Collapse
|
18
|
Baxter AW, Wyllie DJA. Phosphatidylinositol 3 kinase activation and AMPA receptor subunit trafficking underlie the potentiation of miniature EPSC amplitudes triggered by the activation of L-type calcium channels. J Neurosci 2006; 26:5456-69. [PMID: 16707798 PMCID: PMC6675302 DOI: 10.1523/jneurosci.4101-05.2006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We have characterized a mechanism by which the amplitudes of miniature EPSCs (mEPSCs) in CA1 pyramidal neurons in rat hippocampal organotypic slice cultures are potentiated by approximately twofold after a series of depolarizing voltage pulses from -80 to +20 mV. The increase in mEPSC amplitudes is triggered by the activation of L-type calcium channels and is independent of NMDA receptor (NMDAR) activation but also requires calcium release from intracellular stores. The potentiation induced by depolarizing pulses does not alter the kinetic parameters of mEPSCs. The induction phase of this potentiation involves phosphatidylinositol 3 kinase (PI3 kinase) activation because it is blocked completely in the presence of the PI3 kinase inhibitors wortmannin and 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one (LY294002). Furthermore, we show that the maintenance phase of depolarizing pulse potentiation requires continued PI3 kinase activity because the application of either wortmannin or LY294002 results in a reversal to control levels of the amplitudes of mEPSCs. Finally, we demonstrate that the increase in mEPSC amplitudes is mediated by the increased expression of functional AMPA receptors (AMPARs) because the potentiation is blocked by N-ethylmaleimide, botulinum toxin A, and a variety of short-sequence peptides that disrupt the interaction of AMPAR subunits with proteins involved with the trafficking of these to the cell membrane. Our data are consistent with the notion that PI3 kinase and membrane fusion/trafficking events play a pivotal role in coordinating changes in synaptic strength, mediated by AMPA receptors, which are triggered by alterations in postsynaptic calcium concentrations whether these changes are initiated via NMDAR-dependent or NMDAR-independent routes.
Collapse
|
19
|
O'Connor DH, Wittenberg GM, Wang SSH. Dissection of Bidirectional Synaptic Plasticity Into Saturable Unidirectional Processes. J Neurophysiol 2005; 94:1565-73. [PMID: 15800079 DOI: 10.1152/jn.00047.2005] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In populations of synapses, overall synaptic strength can undergo either a net strengthening (long-term potentiation) or weakening (long-term depression). These phenomena have distinct induction pathways, but the functional outcome is usually measured as a single lumped quantity. In hippocampal CA3-CA1 synapses, we took two approaches to study the activity dependence of each phenomenon in isolation. First, we selectively blocked one process by applying kinase or phosphatase inhibitors known, respectively, to block potentiation or depression. Second, we saturated depression or potentiation and examined the activity dependence of the converse process. The resulting unidirectional learning rules could be recombined to give a well-known bidirectional frequency-dependent learning rule under the assumption that when both pathways are activated kinases dominate, resulting in potentiation. Saturation experiments revealed an additional process in which potentiated synapses can be locked at high strength. Saturability of the components of plasticity implies that the amount of plasticity contributed by each pathway depends on the initial level of strength of the synapses. Variation in the distribution of initial synaptic strengths predicts a form of metaplasticity and can account for differences in learning rules observed under several physiological and genetic manipulations.
Collapse
Affiliation(s)
- Daniel H O'Connor
- Department of Molecular Biology and Program in Neuroscience, Princeton University, Princeton, NJ 08544, USA
| | | | | |
Collapse
|
20
|
Kelly PT, Mackinnon RL, Dietz RV, Maher BJ, Wang J. Postsynaptic IP3 receptor-mediated Ca2+ release modulates synaptic transmission in hippocampal neurons. ACTA ACUST UNITED AC 2005; 135:232-48. [PMID: 15857686 DOI: 10.1016/j.molbrainres.2004.12.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2004] [Revised: 12/15/2004] [Accepted: 12/20/2004] [Indexed: 11/16/2022]
Abstract
Ca(2+)-dependent mechanisms are important in regulating synaptic transmission. The results herein indicate that whole-cell perfusion of inositol 1,4,5-trisphosphate receptor (IP(3)R) agonists greatly enhanced excitatory postsynaptic current (EPSC) amplitudes in postsynaptic hippocampal CA1 neurons. IP(3)R agonist-mediated increases in synaptic transmission changed during development and paralleled age-dependent increases in hippocampal type-1 IP(3)Rs. IP(3)R agonist-mediated increases in EPSC amplitudes were inhibited by postsynaptic perfusion of inhibitors of Ca(2+)/calmodulin, PKC and Ca(2+)/calmodulin-dependent protein kinase II. Postsynaptic perfusion of inhibitors of smooth endoplasmic reticulum (SER) Ca(2+)-ATPases, which deplete intracellular Ca(2+) stores, also enhanced EPSC amplitudes. Postsynaptic perfusion of the IP(3)R agonist adenophostin (AdA) during subthreshold stimulation appeared to convert silent to active synapses; synaptic transmission at these active synapses was completely blocked by 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX). Postsynaptic IP(3)R-mediated Ca(2+) release also produced a significant increase in spontaneous EPSC frequency. These results indicate that Ca(2+) release from intracellular stores play a key role in regulating the function of postsynaptic AMPARs.
Collapse
MESH Headings
- 2-Amino-5-phosphonovalerate/pharmacology
- 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology
- Adenosine/analogs & derivatives
- Adenosine/pharmacology
- Age Factors
- Animals
- Animals, Newborn
- Bicuculline/pharmacology
- Blotting, Western/methods
- Calcium/metabolism
- Calcium Channel Agonists/pharmacology
- Calcium Channels/physiology
- Drug Interactions
- Electric Stimulation/methods
- Enzyme Inhibitors/pharmacology
- Excitatory Amino Acid Antagonists/pharmacology
- Excitatory Postsynaptic Potentials/drug effects
- Excitatory Postsynaptic Potentials/physiology
- Excitatory Postsynaptic Potentials/radiation effects
- GABA Antagonists/pharmacology
- Gene Expression Regulation, Developmental/drug effects
- Gene Expression Regulation, Developmental/physiology
- Hippocampus/cytology
- Hippocampus/growth & development
- In Vitro Techniques
- Indoles/pharmacology
- Inositol 1,4,5-Trisphosphate/analogs & derivatives
- Inositol 1,4,5-Trisphosphate/pharmacology
- Inositol 1,4,5-Trisphosphate Receptors
- Neurons/drug effects
- Neurons/physiology
- Neurons/radiation effects
- Patch-Clamp Techniques/methods
- Picrotoxin/pharmacology
- Rats
- Receptors, Cytoplasmic and Nuclear/agonists
- Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors
- Receptors, Cytoplasmic and Nuclear/physiology
- Synaptic Transmission/drug effects
- Synaptic Transmission/physiology
- Thapsigargin/pharmacology
- Time Factors
Collapse
Affiliation(s)
- Paul T Kelly
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045-2106, USA.
| | | | | | | | | |
Collapse
|
21
|
Goforth PB, Ellis EF, Satin LS. Mechanical Injury Modulates AMPA Receptor Kinetics via an NMDA Receptor–Dependent Pathway. J Neurotrauma 2004; 21:719-32. [PMID: 15253800 DOI: 10.1089/0897715041269704] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Alterations in glutamatergic transmission are thought to contribute to secondary neuronal damage following traumatic brain injury. Using an in vitro cell injury model, we previously demonstrated an apparent reduction in AMPA receptor desensitization and resultant potentiation of AMPA-evoked currents after stretch injury of cultured neonatal rat cortical neurons. In the present study, we sought to further characterize injury-induced enhancement of AMPA current and elucidate the mechanisms responsible for this pathological process. Using the patch-clamp technique, agonist-activated currents were recorded from control and injured neurons. Potentiation of AMPA-mediated currents occurred quickly, within 15-30 min following injury, and persisted for at least 24 h. Stretch-injury slowed the activation and desensitization of AMPA mediated currents recorded from excised outside-out patches. The co-application of 100 microM AMPA and 20 microM thiocyanate enhanced AMPA receptor desensitization in control neurons and restored desensitization in injured neurons. The potentiation of AMPA-elicited current was prevented by the NMDA receptor antagonist D-APV (20 microM) or the CaMKII inhibitor KN93 (10 microM). These results suggest that mechanical injury initiates a biochemical cascade that involves NMDA receptor and CaMKII activation and produces a long-lasting reduction of AMPA receptor desensitization, which may contribute to the pathophysiology of traumatic brain injury.
Collapse
Affiliation(s)
- Paulette B Goforth
- Department of Pharmacology and Toxicology, Virginia Commonwealth University Medical Center, Richmond, Virginia 23298, USA
| | | | | |
Collapse
|
22
|
Abstract
The molecular mechanisms of learning and memory, and the underlying bidirectional changes in synaptic plasticity that sustain them largely implicate protein kinases and phosphatases. Specifically, Ca(2+)-dependent kinases and phosphatases actively control neuronal processing by forming a tightly regulated balance in which they oppose each other. In this balance, calcineurin (PP2B) is a critical protein phosphatase whose main function is to negatively modulate learning, memory, and plasticity. It acts by dephosphorylating numerous substrates in different neuronal compartments. This review outlines some of CN neuronal targets and their implication in synaptic functions, and describes the role of CN in the acquisition, storage, retrieval, and extinction of memory, as well as in bidirectional plasticity.
Collapse
Affiliation(s)
- Isabelle M Mansuy
- Department of Biology, Swiss Federal Institute of Technology, ETH Hönggerberg HPM D24, CH-8093, Zurich, Switzerland
| |
Collapse
|
23
|
Ramírez-Munguía N, Vera G, Tapia R. Epilepsy, neurodegeneration, and extracellular glutamate in the hippocampus of awake and anesthetized rats treated with okadaic acid. Neurochem Res 2003; 28:1517-24. [PMID: 14570396 DOI: 10.1023/a:1025670308663] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
We have previously shown that the intrahippocampal microinjection of okadaic acid (OKA), a potent inhibitor of serine/threonine protein phosphatases, induces epileptic seizures, neuronal death, and the hyperphosphorylation of the NR2B subunit of the N-methyl-D-aspartate (NMDA) receptor. We administered OKA by reverse microdialysis in the hippocampus of awake and halothane-anesthetized rats, with simultaneous collection of microdialysis fractions and recording of the EEG activity, and subsequent histological analysis. OKA produced intense behavioral and persistent EEG seizure activity in the awake rats but not in the anesthetized animals, and did not significantly alter the extracellular concentration of glutamate and aspartate detected in the microdialysis fractions. One day after the experiment a remarkable neurodegeneration of CA1 hippocampal region was observed in both the awake and the anesthetized rats. We conclude that the OKA-induced epilepsy cannot be ascribed to increased extracellular glutamate, but to an increased sensitivity of NMDA receptor. We propose that halothane protected against the epilepsy because it blocks NMDA receptor overactivation, and that the neurodegeneration of CA1 region is independent of this overactivation and due probably to alterations of cytoskeletal proteins consequent to the OKA-induced hyperphosphorylation.
Collapse
Affiliation(s)
- Nadia Ramírez-Munguía
- Departamento de Neurociencias, Instituto de Fisiología Celular, Unversidad Nacional Autónoma de México, AP-70-253, 04510-México, DF, México
| | | | | |
Collapse
|
24
|
Hendricson AW, Thomas MP, Lippmann MJ, Morrisett RA. Suppression of L-type voltage-gated calcium channel-dependent synaptic plasticity by ethanol: analysis of miniature synaptic currents and dendritic calcium transients. J Pharmacol Exp Ther 2003; 307:550-8. [PMID: 12970385 DOI: 10.1124/jpet.103.055137] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Intoxicating concentrations of ethanol inhibit N-methyl-d-aspartate (NMDA) receptor-dependent long-term potentiation, an interaction thought to underlie a major component of the central nervous system actions of ethanol. Another form of synaptic potentiation involving activation of L-type dihydropyridine-sensitive voltage-gated calcium channels (VGCCs) has been described, but very little information concerning ethanol effects on VGCC-dependent synaptic potentiation is available. Here, we assessed ethanol effects on VGCC-dependent synaptic potentiation using whole cell patch-clamp recordings of alpha-amino-3-hydroxy-5-methyl-4-soxazolepropionic acid (AMPA) receptor-mediated miniature excitatory postsynaptic currents (mEPSCs) in area CA1 of the rat hippocampus. No potentiation was observed in artificial cerebrospinal fluid containing 2 to 3 mM Ca2+, but marked potentiation of mEPSCs was consistently observed in 4 mM Ca2+ and with patch pipettes containing an ATP-regenerating system. This potentiation was insensitive to the NMDA receptor antagonist DL-2-amino-5-phosphonovaleric acid, whereas it was completely blocked the L-type VGCC antagonist nifedipine. Potentiation was also blocked dose dependently by bath application of ethanol (25-75 mM), which had no effect on baseline mEPSC amplitude or frequency. The synaptic potentiation involved enhancement of both presynaptic and postsynaptic components because significant increases in both the frequency and amplitude of AMPA mEPSCs were observed. Ethanol inhibition of VGCC-dependent synaptic potentiation seemed to occur at the induction step because both the increases in mEPSC frequency and amplitude were affected. To address that question more directly, we used fluorescent imaging of synaptically evoked dendritic calcium events, which displayed a similarly marked ethanol sensitivity. Thus, ethanol modulates fast excitatory synaptic transmission by inhibiting the induction of an NMDA receptor-independent form of synaptic potentiation observed at excitatory synapses on central neurons.
Collapse
Affiliation(s)
- Adam W Hendricson
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, 1 University Station, Austin, TX 78712-0125, USA
| | | | | | | |
Collapse
|
25
|
Cheng G, Randić M. Involvement of intracellular calcium and protein phosphatases in long-term depression of A-fiber-mediated primary afferent neurotransmission. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2003; 144:73-82. [PMID: 12888218 DOI: 10.1016/s0165-3806(03)00161-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Long-term depression (LTD) of monosynaptic and polysynaptic excitatory postsynaptic potentials (EPSPs) in substantia gelatinosa (SG) neurons can be induced by brief high-frequency electrical stimulation (HFS, 300 pulses at 100 Hz) of primary afferent fibers in dorsal roots. Here we examined the possible cellular mechanism underlying spinal LTD. Conventional intracellular recordings were made from SG neurons in a transverse slice-dorsal root preparation of the young rat lumbar spinal cord. LTD of both monosynaptic and polysynaptic EPSPs was induced in 16 of 24 SG neurons by HFS of dorsal root in either the presence or absence of the GABA(A) receptor antagonist bicuculline and the glycine receptor antagonist strychnine. Loading the postsynaptic cell with BAPTA, an intracellular Ca(2+) chelator, almost completely blocked the induction of LTD. Induction of LTD was abolished by bath application of calyculin A (100 nM), a potent inhibitor of protein phosphatases 1 and 2A. These results indicate that: (i) a rise in postsynaptic Ca(2+) is necessary for LTD induction, (ii) synaptic activation of protein phosphatases 1 and 2A plays an important role in the induction of LTD of primary afferent A-fiber neurotransmission in the young rat spinal cord, and (iii) the effect of LTD may be physiologically relevant for transmission and integration of sensory information, including nociception.
Collapse
Affiliation(s)
- Gong Cheng
- Department of Biomedical Sciences, Iowa State University, Ames, IA 50011-1250, USA.
| | | |
Collapse
|
26
|
Thiagarajan TC, Piedras-Renteria ES, Tsien RW. alpha- and betaCaMKII. Inverse regulation by neuronal activity and opposing effects on synaptic strength. Neuron 2002; 36:1103-14. [PMID: 12495625 DOI: 10.1016/s0896-6273(02)01049-8] [Citation(s) in RCA: 235] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
We show that alpha and betaCaMKII are inversely regulated by activity in hippocampal neurons in culture: the alpha/beta ratio shifts toward alpha during increased activity and beta during decreased activity. The swing in ratio is approximately 5-fold and may help tune the CaMKII holoenzyme to changing intensities of Ca(2+) signaling. The regulation of CaMKII levels uses distinguishable pathways, one responsive to NMDA receptor blockade that controls alphaCaMKII alone, the other responsive to AMPA receptor blockade and involving betaCaMKII and possibly further downstream effects of betaCaMKII on alphaCaMKII. Overexpression of alphaCaMKII or betaCaMKII resulted in opposing effects on unitary synaptic strength as well as mEPSC frequency that could account in part for activity-dependent effects observed with chronic blockade of AMPA receptors. Regulation of CaMKII subunit composition may be important for both activity-dependent synaptic homeostasis and plasticity.
Collapse
Affiliation(s)
- Tara C Thiagarajan
- Department of Molecular and Cellular Physiology, Beckman Center, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | |
Collapse
|
27
|
Abstract
Protein dephosphorylation mediated by phosphatases represents an important mechanism for modulating the functions of the targeted proteins. Calyculin A has been extensively used as a specific inhibitor of protein phosphatases. However, the effect of calyculin A on K channel currents in vascular smooth muscle cells (SMCs) and the underlying mechanisms had been unknown. It was found in the current study that calyculin A inhibited the whole-cell outward K channel currents in rat tail artery SMCs in a concentration-dependent (median inhibitory concentration, 12.6 n ) and reversible fashion. The extracellular applied calyculin A induced a biphasic change in K current amplitude with an initial transient increase followed by a long-lasting inhibition (n = 6). The intracellularly applied calyculin A (100 nM ) caused a lesser inhibition (33 +/- 1%) of K channel currents than that caused by the extracellularly applied calyculin A (55.3 +/- 8% inhibition) and did not result in an initial increase in K channel currents. The inhibitory effect of the intracellularly applied calyculin A on K channel currents was reversed to a stimulatory effect after ATP was omitted from the intracellular solution. The K currents inhibited by calyculin A were conducted by the iberiotoxin-sensitive K channels in SMCs. Moreover, okadaic acid (0.03-3 microM ) did not cause any significant change in K(Ca) channel currents. In conclusion, calyculin A inhibited K(Ca) channel currents in vascular SMCs. This effect of calyculin A, however, was not mediated by the inhibition of protein phosphatases.
Collapse
Affiliation(s)
- Salma Toma Hanna
- Department of Physiology, University of Saskatchewan, Saskatoon, SK, Canada
| | | |
Collapse
|
28
|
Semyanov A, Godukhin O. Epileptiform activity and EPSP-spike potentiation induced in rat hippocampal CA1 slices by repeated high-K(+): involvement of ionotropic glutamate receptors and Ca(2+)/calmodulin-dependent protein kinase II. Neuropharmacology 2001; 40:203-11. [PMID: 11114399 DOI: 10.1016/s0028-3908(00)00147-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
We have previously demonstrated that repeated brief increases in extracellular K(+) (K(+)(o)) induce a hyperexcitability in CA1 pyramidal cells that persists for a long time after the final application of K(+) [Neurosci. Lett. 223 (1997) 177; Epilepsy Research (2000) 75]. This epileptiform activity, which was associated with a lasting excitatory postsynaptic potential (EPSP)-spike potentiation, presented some of the characteristic features of traditional in vivo kindling. We have also found that Ca(2+) influx through L-type voltage-sensitive Ca(2+) channels is essential for the development of both in vitro kindling and EPSP-spike potentiation. The aims of this study were to investigate the involvement of ionotropic glutamate receptors, especially those of the NMDA subtype, and the requirement for Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) in these phenomena. Field EPSPs with presynaptic fibre volleys from the stratum radiatum, and population spikes from the stratum pyramidale, were recorded in the CA1 area of rat hippocampal slices in response to electrical stimulation of the Schaffer collateral/commissural fibres. Repeated (three episodes) brief (30 s) increases in extracellular K(+) induced a sustained decrease in the threshold for development of evoked epileptiform discharges (i.e. an in vitro kindling-like state) and a lasting potentiation of the EPSP-spike transfer in CA1 pyramidal neurons (EPSP-spike potentiation). The selective antagonist of NMDA receptors, APV (50 microM), blocked the EPSP-spike potentiation, depressed the induction phase of the in vitro kindling-like state, and blocked the maintenance phase of this state. In contrast to APV, the blockade of AMPA/kainate receptors by CNQX (10 microM) had no effect. Like APV, KN62 (3 microM), a selective membrane permeable inhibitor of CaMKII, blocked the EPSP-spike potentiation and the maintenance phase of the in vitro kindling-like state. Our previous and present results therefore demonstrate that Ca(2+) influxes through L-type voltage-dependent-and NMDA receptor-dependent-Ca(2+) channels contribute differentially to the development of an in vitro kindling-like state, and both induce EPSP-spike potentiation in CA1 hippocampal pyramidal cells in response to repeated brief increases in K(+)(o). It is suggested that these effects of intracellular Ca(2+) on the maintenance phase of the in vitro kindling-like state and EPSP-spike potentiation are mediated by CaMKII-dependent mechanisms.
Collapse
Affiliation(s)
- A Semyanov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142292, Russia.
| | | |
Collapse
|
29
|
Hirai H, Yoshioka K, Yamada K. A simple method using 31P-NMR spectroscopy for the study of protein phosphorylation. BRAIN RESEARCH. BRAIN RESEARCH PROTOCOLS 2000; 5:182-9. [PMID: 10775839 DOI: 10.1016/s1385-299x(00)00011-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Nonradioactive 31P-NMR spectroscopy has previously been used for the study of protein phosphorylations. However, the procedures does not seem to be easy for non-experts of this field, hence, this approach has not been widely used. We introduce here a simple protocol with 31P-NMR spectroscopy to study in vitro phosphorylation in receptor proteins. The effectiveness of this method was verified using synthetic peptides and recombinant proteins of the C-terminus of the alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate (AMPA) receptor, whose phosphorylations are considered to have important roles in synaptic plasticity. We show that a decrease in the pH of the sample solution after the phosphorylation reaction is critical for the separation of the phosphorylation signals. In the analysis of the C-terminal portion of the GluR2 AMPA receptor, the phosphorylation sites of which had not hitherto been well clarified, we found the presence of at least three protein kinase C (PKC) phosphorylation sites. Furthermore, this method allows prediction of the origins of each of the phosphorylation peaks. Thus, the techniques we described here is useful for examination of protein phosphorylation and permits us to safely conduct repetitive experiments.
Collapse
Affiliation(s)
- H Hirai
- Laboratory for Memory and Learning, RIKEN Brain Science Institute, Wako, Japan.
| | | | | |
Collapse
|
30
|
Lu YM, Mansuy IM, Kandel ER, Roder J. Calcineurin-mediated LTD of GABAergic inhibition underlies the increased excitability of CA1 neurons associated with LTP. Neuron 2000; 26:197-205. [PMID: 10798404 DOI: 10.1016/s0896-6273(00)81150-2] [Citation(s) in RCA: 219] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Coincident pre- and postsynaptic activity generates long-term potentiation (LTP), a possible cellular model of learning and memory. LTP has two components: (1) an increase in the excitatory postsynaptic potential (EPSP), and (2) an increase in the ability of the EPSP to generate a spike (E-S coupling of LTP). We have used pharmacological and genetic approaches to address the molecular nature of E-S coupling in CA1 pyramidal neurons. Blockade of the Ca2+-sensitive phosphatase, calcineurin, prevents induction of E-S coupling without interfering with LTP of the EPSP. Calcineurin produces its effect on E-S coupling by inducing a long-lasting depression (LTD) of the GABA(A)-mediated inhibitory postsynaptic potentials (IPSPs). This LTD of the IPSP was prevented by blockade of NMDA receptors. Thus, the tetanus that elicits NMDA-dependent LTP mediates a coordinately regulated double function. It produces LTP of the EPSP and, concomitantly, LTD of the IPSP that leads to enhancement of E-S coupling.
Collapse
Affiliation(s)
- Y M Lu
- Department of Molecular and Medical Genetics, University of Toronto, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Canada
| | | | | | | |
Collapse
|
31
|
Mehler MF. Brain dystrophin, neurogenetics and mental retardation. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 2000; 32:277-307. [PMID: 10751678 DOI: 10.1016/s0165-0173(99)00090-9] [Citation(s) in RCA: 134] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Duchenne muscular dystrophy (DMD) and the allelic disorder Becker muscular dystrophy (BMD) are common X-linked recessive neuromuscular disorders that are associated with a spectrum of genetically based developmental cognitive and behavioral disabilities. Seven promoters scattered throughout the huge DMD/BMD gene locus normally code for distinct isoforms of the gene product, dystrophin, that exhibit nervous system developmental, regional and cell-type specificity. Dystrophin is a complex plasmalemmal-cytoskeletal linker protein that possesses multiple functional domains, autosomal and X-linked homologs and associated binding proteins that form multiunit signaling complexes whose composition is unique to each cellular and developmental context. Through additional interactions with a variety of proteins of the extracellular matrix, plasma membrane, cytoskeleton and distinct intracellular compartments, brain dystrophin acquires the capability to participate in the modulatory actions of a large number of cellular signaling pathways. During neural development, dystrophin is expressed within the neural tube and selected areas of the embryonic and postnatal neuraxis, and may regulate distinct aspects of neurogenesis, neuronal migration and cellular differentiation. By contrast, in the mature brain, dystrophin is preferentially expressed by specific regional neuronal subpopulations within proximal somadendritic microdomains associated with synaptic terminal membranes. Increasing experimental evidence suggests that in adult life, dystrophin normally modulates synaptic terminal integrity, distinct forms of synaptic plasticity and regional cellular signal integration. At a systems level, dystrophin may regulate essential components of an integrated sensorimotor attentional network. Dystrophin deficiency in DMD/BMD patients and in the mdx mouse model appears to impair intracellular calcium homeostasis and to disrupt multiple protein-protein interactions that normally promote information transfer and signal integration from the extracellular environment to the nucleus within regulated microdomains. In DMD/BMD, the individual profiles of cognitive and behavioral deficits, mental retardation and other phenotypic variations appear to depend on complex profiles of transcriptional regulation associated with individual dystrophin mutations that result in the corresponding presence or absence of individual brain dystrophin isoforms that normally exhibit developmental, regional and cell-type-specific expression and functional regulation. This composite experimental model will allow fine-level mapping of cognitive-neurogenetic associations that encompass the interrelationships between molecular, cellular and systems levels of signal integration, and will further our understanding of complex gene-environmental interactions and the pathogenetic basis of developmental disorders associated with mental retardation.
Collapse
Affiliation(s)
- M F Mehler
- Departments of Neurology, Neuroscience and Psychiatry, the Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
32
|
Moon C, Fraser SP, Djamgoz MB. Protein kinase and phosphatase modulation of quail brain GABA(A) and non-NMDA receptors co-expressed in Xenopus oocytes. Cell Signal 2000; 12:105-12. [PMID: 10679579 DOI: 10.1016/s0898-6568(99)00073-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The GABA(A) receptor and the non-NMDA subtype of the ionotropic glutamate receptor were co-expressed in Xenopus oocytes by injection of quail brain mRNA. The oocytes were treated with various protein kinase (PK) and protein phosphatase (PP) activators and inhibitors and the effects on receptor functioning were monitored. Two phorbol esters, 4-beta-phorbol 12-myristate-13-acetate (PMA) and 4-beta-phorbol 12,13-dibutyrate (PDBu); the cGMP-dependent PK activators sodium nitroprusside (SNP) and S-nitrosoglutathione (SNOG); and the PP inhibitor okadaic acid (OA) reduced the amplitude of the GABA-induced currents, whilst the PK inhibitor staurosporine potentiated it. In addition, PMA, PDBu, SNP, and OA reduced the desensitization of the GABA-induced response. Identical treatments generally had similar but less pronounced effects on responses generated by kainate (KA) but the desensitization characteristic of the non-NMDA receptor was not affected. None of the treatments had any effect on the reversal potentials of the induced currents. Immunoblots revealed that the oocytes express endogenous PKG and guanylate cyclase. The results are discussed in terms of the molecular structures of GABA(A) and non-NMDA receptors and the potential functional consequences of phosphorylation/dephosphorylation.
Collapse
Affiliation(s)
- C Moon
- Neurobiology Group, Department of Biology, Imperial College of Science, Technology and Medicine, Sir Alexander Fleming Building, London, UK
| | | | | |
Collapse
|
33
|
Herzig S, Neumann J. Effects of serine/threonine protein phosphatases on ion channels in excitable membranes. Physiol Rev 2000; 80:173-210. [PMID: 10617768 DOI: 10.1152/physrev.2000.80.1.173] [Citation(s) in RCA: 202] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This review deals with the influence of serine/threonine-specific protein phosphatases on the function of ion channels in the plasma membrane of excitable tissues. Particular focus is given to developments of the past decade. Most of the electrophysiological experiments have been performed with protein phosphatase inhibitors. Therefore, a synopsis is required incorporating issues from biochemistry, pharmacology, and electrophysiology. First, we summarize the structural and biochemical properties of protein phosphatase (types 1, 2A, 2B, 2C, and 3-7) catalytic subunits and their regulatory subunits. Then the available pharmacological tools (protein inhibitors, nonprotein inhibitors, and activators) are introduced. The use of these inhibitors is discussed based on their biochemical selectivity and a number of methodological caveats. The next section reviews the effects of these tools on various classes of ion channels (i.e., voltage-gated Ca(2+) and Na(+) channels, various K(+) channels, ligand-gated channels, and anion channels). We delineate in which cases a direct interaction between a protein phosphatase and a given channel has been proven and where a more complex regulation is likely involved. Finally, we present ideas for future research and possible pathophysiological implications.
Collapse
Affiliation(s)
- S Herzig
- Institut für Pharmakologie, Universität Köln, Köln, Germany.
| | | |
Collapse
|
34
|
Foster TC. Involvement of hippocampal synaptic plasticity in age-related memory decline. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 1999; 30:236-49. [PMID: 10567726 DOI: 10.1016/s0165-0173(99)00017-x] [Citation(s) in RCA: 239] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
This article examines the functional significance of Ca(2+)-dependent synaptic plasticity in relation to compromised memory function during aging. Research characterizing an age-related decline in memory for tasks that require proper hippocampal function is summarized. It is concluded that aged animals possess the mechanisms necessary for memory formation, and memory deficits, including rapid forgetting, result from more subtle changes in memory processes for memory storage or maintenance. A review of experimental studies concerning changes in hippocampal neural plasticity over the course of aging indicates that, during aging, there is a shift in mechanisms that regulate the thresholds for synaptic modification, including Ca(2+) channel function and subsequent Ca(2+)-dependent processes. The results, combined with theoretical considerations concerning synaptic modification thresholds, provide the basis for a model of age-related changes in hippocampal synaptic function. The model is employed as a foundation for interpretation of studies examining therapeutic intervention in age-related memory decline. The possible role of altered synaptic plasticity thresholds in learning and memory deficits suggests that treatments that modify synaptic plasticity may prove fruitful for the development of early therapeutic interventions in age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- T C Foster
- Department of Pharmacology, College of Medicine, University of Kentucky, MS-305 UKMC, Lexington, KY, USA.
| |
Collapse
|
35
|
Tapia R, Peña F, Arias C. Neurotoxic and synaptic effects of okadaic acid, an inhibitor of protein phosphatases. Neurochem Res 1999; 24:1423-30. [PMID: 10555783 DOI: 10.1023/a:1022588808260] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Protein phosphorylation and dephosphorylation reactions, catalyzed by kinases and phosphatases, are involved in the regulation of a wide variety of physiological processes. In the nervous system, such reactions seem to modulate the function of several proteins crucial in synaptic transmission, including voltage-gated and ligand-gated channels, neurotransmitter release, and neurotransmitter transporters. On the other hand, hyperphosphorylation of certain cytoskeletal proteins or receptors may lead to neuronal death. In the present work we review the neurotoxic effect of okadaic acid (OKA), a potent and specific inhibitor of the serine/threonine protein phosphatases 1 and 2A, as well as its action on synaptic function. We analyze recent findings demonstrating that the microinjection of OKA in rat hippocampus induces neuronal stress, hyperexcitation and neurodegeneration, and discuss their possible relationships to alterations of protein phosphorylation-dephosphorylation observed in Alzheimer's disease brain. These results suggest that protein hyperphosphorylation due to inhibition of phosphatases in vivo induces neuronal stress and subsequent neurodegeneration.
Collapse
Affiliation(s)
- R Tapia
- Departamento de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, DF, México.
| | | | | |
Collapse
|
36
|
Stricker C, Cowan AI, Field AC, Redman SJ. Analysis of NMDA-independent long-term potentiation induced at CA3-CA1 synapses in rat hippocampus in vitro. J Physiol 1999; 520 Pt 2:513-25. [PMID: 10523419 PMCID: PMC2269604 DOI: 10.1111/j.1469-7793.1999.00513.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
1. Excitatory postsynaptic currents (EPSCs) were evoked at synapses formed by Schaffer collaterals/commissural (CA3) axons with CA1 pyramidal cells using the rat hippocampal slice preparation. Long-term potentiation (LTP) was induced at these synapses using a pairing protocol, with 50 microM d,l-APV present in the artificial cerebrospinal fluid (ACSF). 2. Quantal analysis of the amplitudes of the control and conditioned EPSCs showed that the enhancement of synaptic strength was due entirely to an increase in quantal content of the EPSC. No change occurred in the quantal current. 3. These results were compared with those obtained from a previous quantal analysis of LTP induced in normal ACSF, where both quantal current and quantal content increased. The results suggest that calcium entering via NMDA receptors initiates the signalling cascade that results in enhanced AMPA currents because it is adding to cytoplasmic calcium from other sources to reach a threshold for this signalling pathway, or because calcium entering via NMDA receptors specifically activates this signalling pathway.
Collapse
Affiliation(s)
- C Stricker
- Division of Neuroscience, John Curtin School of Medical Research, Australian National University, Institute of Neuroinformatics, Zurich, Switzerland
| | | | | | | |
Collapse
|
37
|
Zwick E, Wallasch C, Daub H, Ullrich A. Distinct calcium-dependent pathways of epidermal growth factor receptor transactivation and PYK2 tyrosine phosphorylation in PC12 cells. J Biol Chem 1999; 274:20989-96. [PMID: 10409647 DOI: 10.1074/jbc.274.30.20989] [Citation(s) in RCA: 92] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Recently, we have demonstrated that in PC12 cells activation of the Ras/extracellular signal-regulated kinase pathway in response to membrane depolarization or bradykinin is mediated by calcium-dependent transactivation of the epidermal growth factor receptor (EGFR). Here we address the question whether Ca(2+)-calmodulin-dependent protein kinase (CaM kinase) has a role in the EGFR transactivation signal. Using compounds that selectively interfere with either CaM kinase activity or calmodulin function, we show that KCl-mediated membrane depolarization-triggered, but not bradykinin-mediated signals involve CaM kinase function upstream of the EGFR. Although both depolarization-induced calcium influx and bradykinin stimulation of PC12 cells were found to induce c-fos transcription through EGFR activation, the former signal is CaM kinase-dependent and the latter was shown to be independent. As PYK2 is also activated upon elevation of intracellular calcium, we investigated the potential involvement of this cytoplasmic tyrosine kinase in EGFR transactivation. Interestingly, we observed that inhibition of CaM kinase activity in PC12 cells abrogated tyrosine phosphorylation of PYK2 upon KCl but not bradykinin treatment. Nevertheless, PYK2 activation in response to both stimuli appeared to be mediated by pathways parallel to EGFR transactivation. Our data demonstrate the existence of two distinct calcium-dependent mechanisms leading either to EGFR-mediated extracellular signal-regulated activation or to PYK2 tyrosine phosphorylation. Both pathways either in concert or independently might contribute to the definition of biological responses in neuronal cell types.
Collapse
Affiliation(s)
- E Zwick
- Department of Molecular Biology, Max-Planck-Institut für Biochemie, Am Klopferspitz 18A, 82152 Martinsried, Germany
| | | | | | | |
Collapse
|
38
|
Ahn S, Ginty DD, Linden DJ. A late phase of cerebellar long-term depression requires activation of CaMKIV and CREB. Neuron 1999; 23:559-68. [PMID: 10433267 DOI: 10.1016/s0896-6273(00)80808-9] [Citation(s) in RCA: 146] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Recently, it has been shown that cerebellar LTD has a late phase that may be blocked by protein synthesis inhibitors. To understand the mechanisms underlying the late phase, we interfered with the activation of transcription factors that might couple synaptic activation to protein synthesis. Particle-mediated transfection of cultured Purkinje neurons with an expression vector encoding a dominant inhibitory form of CREB resulted in a nearly complete blockade of the late phase. Kinases that activate CREB were inhibited, and LTD was assessed. Inhibition of PKA or the MAPK/RSK cascades were without effect on the late phase, while constructs designed to interfere with CaMKIV function attenuated the late phase. These results indicate that the activation of CaMKIV and CREB are necessary to establish a late phase of cerebellar LTD.
Collapse
Affiliation(s)
- S Ahn
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | |
Collapse
|
39
|
Ezrokhi VL, Zosimovskii VA, Korshunov VA, Markevich VA. Restoration of decaying long-term potentiation in the hippocampal formation by stimulation of neuromodulatory nuclei in freely moving rats. Neuroscience 1999; 88:741-53. [PMID: 10363814 DOI: 10.1016/s0306-4522(98)00232-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Induction of long-term potentiation within the hippocampal formation can be modulated by afferent influences from a number of subcortical structures known to be involved in hippocampal-dependent learning and memory. This study performed on freely moving rats investigated the effects of stimulation of the noradrenergic locus coeruleus nucleus and the serotonergic dorsal raphe nucleus on spontaneously decaying posttetanic long-term potentiation in the dentate gyrus and the hippocampal CA1 area, respectively. High-frequency electrical stimulation of the locus coeruleus or the dorsal raphe elicited a well-expressed behavioural reaction of exploratory or defensive type, respectively, but did not significantly alter transmission at perforant path-dentate gyrus or Schaffer collateral-CA synapses, when delivered either before tetanic stimulation of the perforant path or the Schaffer collaterals or long (hours and days) after previously induced long-term potentiation had completely decayed. However, when locus coeruleus or dorsal raphe stimulation was delivered with the same parameters during a limited time (minutes and hours) after marked or even complete decay of tetanus-induced long-term potentiation at perforant path-dentate gyrus or Schaffer collateral-CA1 synapses, the potentiation was partially or entirely restored but never increased beyond the initial level of potentiation. In CA1, stimulation of ipsilateral and contralateral Schaffer collaterals demonstrated that the restoration of previously existing long-term potentiation by dorsal raphe stimulation was input-specific, occurring, like tetanus-induced potentiation, only in the pathway which had previously been tetanized. These findings suggest that the noradrenergic locus coeruleus and the serotonergic dorsal raphe can influence not only induction, but also spontaneous decay of long-term potentiation in the hippocampal formation. Since hippocampal long-term potentiation is thought to play a role in certain kinds of learning and memory, and association of tetanic stimulation with activation of ascending neuromodulatory systems is required for full expression of long-term potentiation, the restoration of hippocampal long-term potentiation by activation of a neuromodulatory system alone may serve as a mechanism of associative reminder which may underlie facilitation of memory retrieval after a period of forgetting, as has been observed in trained rats under similar conditions.
Collapse
Affiliation(s)
- V L Ezrokhi
- Laboratory of Neurophysiology of Learning, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow
| | | | | | | |
Collapse
|
40
|
Abstract
Antibodies directed against the mammalian alpha and beta subunits of calcium/calmodulin-dependent kinase 2 (CaMK2) and brain dissection were used for immunoblot analysis of these proteins in various brain regions of Apteronotus leptorhynchus. Western blots revealed that the CaMK2alpha antibody labeled a single band of the expected molecular mass (approximately 50 kDa) for this enzyme in rat cortex and electric fish brain. CaMK2alpha was enriched in fish forebrain and hypothalamus and also strongly expressed in midbrain sensory areas. Western blots revealed that CaMK2beta antibodies labeled bands in an appropriate molecular mass range (approximately 58-64 kDa) for this enzyme in mammalian cortex and electric fish brain. However, a higher molecular mass band (approximately 80 kDa) was also labeled; because all these bands were eliminated by preadsorbtion with the CaMK2-derived peptide antigen, they may all represent CaMK2beta-like isoforms. We mapped the brain distribution of CaMK2 isoforms with emphasis on the electrosensory system. CaMK2alpha was present at high density in dorsal forebrain, hypothalamic nuclei, torus semicircularis, and tectum. It was also enriched in discrete fiber tracts in forebrain, diencephalon, and rhombencephalon. CaMK2beta-like isoforms were enriched in ventral forebrain, hypothalamic nuclei, torus semicircularis and the reticular formation. Unlike CaMK2alpha, CaMK2beta -like isoforms were predominantly present in cell bodies and rarely found in fiber tracts or neuropil. In the electrosensory lateral line lobe, CaMK2alpha was restricted to specific feedback fibers, i.e., tractus stratum fibrosum and its terminal field in the ventral molecular layer. In contrast, CaMK2beta-like isoforms were enriched in somata and dendrites of pyramidal cells and granular interneurons.
Collapse
Affiliation(s)
- L Maler
- Department of Cellular and Molecular Medicine, University of Ottawa, Ontario, Canada.
| | | |
Collapse
|
41
|
Abstract
Long-term potentiation (LTP) is currently the best available cellular model for learning and memory in the mammalian brain. In the CA1 region of the hippocampus, as well as in many other areas of the CNS, its induction requires a rise in postsynaptic Ca2+ via activation of NMDA receptors. What happens after the rise in postsynaptic Ca2+ is less clear. This paper summarizes experiments performed over the last decade in slice preparations that address the site of expression of LTP. While a large number of laboratories have contributed importantly to this issue, this review will rely primarily on experiments performed in the authors' laboratory. The experiments to be discussed can be broadly divided into two groups: those designed to determine if an increase in glutamate release occurs during LTP and those designed to determine if a change in postsynaptic sensitivity to glutamate occurs during LTP. Experiments in the first category include the analysis of dual-component excitatory postsynaptic currents (EPSCs), paired-pulse facilitation, saturating release probability, the use of MK-801 to measure release probability, and glial glutamate transporter currents to measure directly the synaptic release of glutamate. Experiments in the second category include analysis of miniature EPSC amplitudes, measurements of synaptic potency, the consequences of loading cells with the constitutively activated form of CaM kinase II, and the evidence that during LTP postsynaptically silent synapses become functional. We will argue that, while numerous experiments fail to support a presynaptic expression mechanism, many experiments do point to a postsynaptic expression mechanism. The decrease in synaptic failures during LTP, the only generally accepted experimental result that supports a presynaptic expression mechanism, can be explained by postsynaptically silent synapses. Future directions for research in this field include activity-dependent targeting of glutamate receptors and the functional consequences of phosphorylation of AMPA receptors.
Collapse
Affiliation(s)
- R A Nicoll
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco 94143-0450, USA.
| | | |
Collapse
|
42
|
Swope SL, Moss SJ, Raymond LA, Huganir RL. Regulation of ligand-gated ion channels by protein phosphorylation. ADVANCES IN SECOND MESSENGER AND PHOSPHOPROTEIN RESEARCH 1999; 33:49-78. [PMID: 10218114 DOI: 10.1016/s1040-7952(99)80005-6] [Citation(s) in RCA: 254] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The studies discussed in this review demonstrate that phosphorylation is an important mechanism for the regulation of ligand-gated ion channels. Structurally, ligand-gated ion channels are heteromeric proteins comprised of homologous subunits. For both the AChR and the GABA(A) receptor, each subunit has a large extracellular N-terminal domain, four transmembrane domains, a large intracellular loop between transmembrane domains M3 and M4, and an extracellular C-terminal domain (Fig. 1B). All the phosphorylation sites on these receptors have been mapped to the major intracellular loop between M3 and M4 (Table 1). In contrast, glutamate receptors appear to have a very large extracellular N-terminal domain, one membrane hairpin loop, three transmembrane domains, a large extracellular loop between transmembrane domains M3 and M4, and an intracellular C-terminal domain (Fig. 1C). Most phosphorylation sites on glutamate receptors have been shown to be on the intracellular C-terminal domain, although some have been suggested to be on the putative extracellular loop between M3 and M4 (Table 1). A variety of extracellular factors and intracellular signal transduction cascades are involved in regulating phosphorylation of these ligand-gated ion channels (Fig. 2). Once again, the AChR at the neuromuscular junction is the most fully understood system. Phosphorylation of the AChR by PKA is stimulated synaptically by the neuropeptide CGRP and in an autocrine fashion by adenosine released from the muscle in response to acetylcholine. In addition, acetylcholine, via calcium influx through the AChR, appears to activate calcium-dependent kinases including PKC to stimulate serine phosphorylation of the receptor. Presently, agrin is the only extracellular factor known to stimulate phosphorylation of the AChR on tyrosine residues. For glutamate receptors, non-NMDA receptor phosphorylation by PKA is stimulated by dopamine, while NMDA receptor phosphorylation by PKA and PKC can be induced via the activation of beta-adrenergic receptors, and metabotropic glutamate or opioid receptors, respectively. In addition, Ca2+ influx through the NMDA receptor has been shown to activate PKC. CaMKII, and calcineurin, resulting in phosphorylation of AMPA receptors (by CaMKII) and inactivation of NMDA receptors (at least in part through calcineurin). In contrast to the AChR and glutamate receptors, no information is presently available regarding the identities of the extracellular factors and intracellular signal transduction cascades that regulate phosphorylation of the GABA(A) receptor. Surely, future studies will be aimed at further clarifying the molecular mechanisms by which the central receptors are regulated. The presently understood functional effects of ligand-gated ion channel phosphorylation are diverse. At the neuromuscular junction, a regulation of the AChR desensitization rate by both serine and tyrosine phosphorylation has been demonstrated. In addition, tyrosine phosphorylation of the AChR or other synaptic components appears to play a role in AChR clustering during synaptogenesis. For the GABA(A) receptor, the data are complex. Both activation and inhibition of GABA(A) receptor currents as a result of PKA and PKC phosphorylation have been reported, while phosphorylation by PTK enhances function. The predominant effect of glutamate receptor phosphorylation by a variety of kinases is a potentiation of the peak current response. However, PKC also modulates clustering of NMDA receptors. This complexity in the regulation of ligand-gated ion channels by phosphorylation provides diverse mechanisms for mediating synaptic plasticity. In fact, accumulating evidence supports the involvement of protein phosphorylation and dephosphorylation of AMPA receptors in LTP and LTD respectively. There has been a dramatic increase in our understanding of the nature by which phosphorylation regulates ligand-gated ion channels. However, many questions remain unanswered. (AB
Collapse
Affiliation(s)
- S L Swope
- Department of Neurology, Georgetown Institute for Cognitive and Computational Neuroscience, Georgetown University Medical Center, Washington, DC 20007, USA
| | | | | | | |
Collapse
|
43
|
Adenylyl cyclase activation modulates activity-dependent changes in synaptic strength and Ca2+/calmodulin-dependent kinase II autophosphorylation. J Neurosci 1999. [PMID: 10087064 DOI: 10.1523/jneurosci.19-07-02500.1999] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Activation of the Ca2+- and calmodulin-dependent protein kinase II (CaMKII) and its conversion into a persistently activated form by autophosphorylation are thought to be crucial events underlying the induction of long-term potentiation (LTP) by increases in postsynaptic Ca2+. Because increases in Ca2+ can also activate protein phosphatases that oppose persistent CaMKII activation, LTP induction may also require activation of signaling pathways that suppress protein phosphatase activation. Because the adenylyl cyclase (AC)-protein kinase A signaling pathway may provide a mechanism for suppressing protein phosphatase activation, we investigated the effects of AC activators on activity-dependent changes in synaptic strength and on levels of autophosphorylated alphaCaMKII (Thr286). In the CA1 region of hippocampal slices, briefly elevating extracellular Ca2+ induced an activity-dependent, transient potentiation of synaptic transmission that could be converted into a persistent potentiation by the addition of phosphatase inhibitors or AC activators. To examine activity-dependent changes in alphaCaMKII autophosphorylation, we replaced electrical presynaptic fiber stimulation with an increase in extracellular K+ to achieve a more global synaptic activation during perfusion of high Ca2+ solutions. In the presence of the AC activator forskolin or the protein phosphatase inhibitor calyculin A, this treatment induced a LTP-like synaptic potentiation and a persistent increase in autophosphorylated alphaCaMKII levels. In the absence of forskolin or calyculin A, it had no lasting effect on synaptic strength and induced a persistent decrease in autophosphorylated alphaCaMKII levels. Our results suggest that AC activation facilitates LTP induction by suppressing protein phosphatases and enabling a persistent increase in the levels of autophosphorylated CaMKII.
Collapse
|
44
|
Xu TL, Li JS, Jin YH, Akaike N. Modulation of the glycine response by Ca2+-permeable AMPA receptors in rat spinal neurones. J Physiol 1999; 514 ( Pt 3):701-11. [PMID: 9882741 PMCID: PMC2269109 DOI: 10.1111/j.1469-7793.1999.701ad.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
1. In acutely isolated rat sacral dorsal commisural nucleus (SDCN) neurones, application of kainate (KA) reversibly potentiated glycine-evoked Cl- currents (IGly) in a concentration-dependent manner. 2. The cellular events underlying the interaction between non-NMDA receptors and glycine receptors were studied by using nystatin-perforated patch and cell-attached single-channel recording modes. 3. The action of KA was not accompanied by a shift in the reversal potential for IGly. In dose-response curves, KA potentiated IGly without significantly changing glycine binding affinity. 4. GYKI 52466 blocked while NS-102 had no effect on the KA-induced potentiation of IGly. 5. The potentiation was reduced when KA was applied in a Ca2+-free extracellular solution or in the presence of BAPTA AM, and was independent of the activation of voltage-dependent Ca2+ channels. 6. Pretreatment with KN-62, a selective Ca2+-calmodulin-dependent protein kinase II (CaMKII) inhibitor, abolished the action of KA. Inhibition of calcineurin converted the KA-induced potentiation to a sustained one. 7. Single-channel recordings revealed that KA decreased the mean closing time of glycine-gated single-channel activity, resulting in an increase in the probability of channel opening. 8. It is proposed that Ca2+ entry through AMPA receptors modulates the glycine receptor function via coactivation of CaMKII and calcineurin in SDCN neurones. This interaction may provide a new postsynaptic mechanism for control of inhibitory synaptic signalling and represent one of the important regulatory mechanisms of spinal nociception.
Collapse
Affiliation(s)
- T L Xu
- Department of Physiology, Faculty of Medicine, Kyushu University, Fukuoka 812-8582, Japan.
| | | | | | | |
Collapse
|
45
|
Abstract
A major effort in neuroscience is directed towards understanding the roles of Ca2+ signalling in the induction of synaptic plasticity. Here, we summarize the evidence concerning Ca2+ signalling, paying particular attention to CA1 excitatory synapses, and its relationship to the induction of long-term potentiation and long-term depression. We discuss the ways in which synaptic activation can elevate Ca2+ postsynaptically and how dendritic spines may act as a Ca2+ compartment which can both isolate and integrate Ca2+ signals.
Collapse
Affiliation(s)
- R Chittajallu
- Department of Anatomy, Medical School, University of Bristol, UK.
| | | | | |
Collapse
|
46
|
Carroll RC, Nicoll RA, Malenka RC. Effects of PKA and PKC on miniature excitatory postsynaptic currents in CA1 pyramidal cells. J Neurophysiol 1998; 80:2797-800. [PMID: 9819284 DOI: 10.1152/jn.1998.80.5.2797] [Citation(s) in RCA: 76] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Protein kinases play an important role in controlling synaptic strength at excitatory synapses on CA1 pyramidal cells. We examined the effects of activating cAMP-dependent protein kinase or protein kinase C (PKC) on the frequency and amplitude of miniature excitatory postsynaptic currents (mEPSCs) with perforated patch recording techniques. Both forskolin and phorbol-12,13-dibutryate (PDBu) caused a large increase in mEPSC frequency, but only PDBu increased mEPSC amplitude, an effect that was not observed when standard whole cell recording was performed. These results support biochemical observations indicating that PKC, similar to calcium/calmodulin-dependent protein kinase II, has an important role in controlling synaptic strength via modulation of AMPA receptor function, potentially through the direct phosphorylation of the GluR1 subunit.
Collapse
Affiliation(s)
- R C Carroll
- Department of Psychiatry, University of California, San Francisco, California 94143, USA
| | | | | |
Collapse
|
47
|
Abstract
Casein kinase 2 is present in the brain, including the hippocampus. It is associated with long-term potentiation and is known to be involved in phosphorylation of proteins potentially important for neuroplasticity, but regulation of its activity in neuronal cells is not yet known. In the present work, it was found that brain-derived neurotrophic factor and neurotrophin-4 control the activity of casein kinase 2 in hippocampal slices of adult rat. It is shown that: (i) treatment of slices for 4 h with the neurotrophins results in a five-fold increase in the activity of cytosolic casein kinase 2; (ii) this effect does not require protein synthesis. In addition, using calcium chelators, phospholipase inhibitors and protein kinase inhibitors, evidence is provided that: (i) neurotrophin-induced activation of casein kinase 2 is dependent on the availability of intracellular calcium due to stimulation of phospholipase C; (ii) both a tyrosine kinase(s) and a serine/threonine kinase(s) convey the signal of calcium. Since there is now accumulating evidence for involvement of brain-derived neurotrophic factor, intracellular calcium, tyrosine kinases and serine/threonine kinases in the regulation of synaptic plasticity, it is suggested that the signalling cascade detected here might contribute to control of synaptic strength in the hippocampus.
Collapse
Affiliation(s)
- P R Blanquet
- Unité de Recherche de Physiopharmacologie du Système Nerveux, U-161 INSERM, Paris, France
| |
Collapse
|
48
|
Thiels E, Norman ED, Barrionuevo G, Klann E. Transient and persistent increases in protein phosphatase activity during long-term depression in the adult hippocampus in vivo. Neuroscience 1998; 86:1023-9. [PMID: 9697109 DOI: 10.1016/s0306-4522(98)00135-3] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The neural substrates of learning and memory most likely involve activity-dependent long-term changes in synaptic strength, including long-term potentiation and long-term depression. A critical element in the cascade of events hypothesized to underlie such changes in synaptic function is modification of protein phosphorylation. Long-term depression is thought to involve decreases in protein phosphorylation, which could result from reduction in protein kinase activity and/or enhancement in protein phosphatase activity. We present here direct evidence that long-term depression in the hippocampus in vivo is associated with an increase in the activity of the serine/threonine phosphatases 1 and 2A. The increase in activity of phosphatase 1 was transient, whereas that of phosphatase 2A lasted > 65 min after the induction of long-term depression. Blockade of long-term depression prevented the observed increases in phosphatase activity, as did selective inhibition of phosphatase 1 and 2A. Induction of long-term depression had no effect on the level of either phosphatase, which suggests that our results reflect increases in the intrinsic activity of these two enzymes. Our findings are consistent with a model of synaptic plasticity that implicates protein dephosphorylation by serine/threonine phosphatases in the early maintenance and/or expression of long-term depression of synaptic strength.
Collapse
Affiliation(s)
- E Thiels
- Department of Neuroscience, University of Pittsburgh, PA 15260, USA
| | | | | | | |
Collapse
|
49
|
Chen HX, Hanse E, Pananceau M, Gustafsson B. Distinct expressions for synaptic potentiation induced by calcium through voltage-gated calcium and N-methyl-D-aspartate receptor channels in the hippocampal CA1 region. Neuroscience 1998; 86:415-22. [PMID: 9881856 DOI: 10.1016/s0306-4522(98)00042-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Brief elevation in postsynaptic calcium in hippocampal CA1 neurons leads to prolonged changes in synaptic strength. The calcium may enter the postsynaptic neuron via different routes, such as voltage-gated calcium channels or glutamate receptor channels of N-methyl-D-aspartate type, and/or be released from intracellular stores. The manner in which the synapse is altered, leading to the expression of an enhanced/depressed synaptic strength, is still unclear. The present study, performed using whole-cell recording from CA1 pyramidal cells of three- to five-week-old guinea-pigs, shows that postsynaptic depolarization alone, allowing for calcium influx through voltage-gated calcium channels, leads to a synaptic potentiation characterized by an altered time-course of the evoked excitatory synaptic response, an unaltered coefficient of variation of that response and a decreased paired-pulse facilitation likely related to a postsynaptic mechanism. These characteristics contrasted with those of long-term potentiation induced via activation of N-methyl-D-aspartate receptor channels, where the time-course was unaltered, the coefficient of variation was decreased and no change in paired-pulse facilitation was observed. Synapses can thus have mechanistically separate, but co-existent, potentiations of synaptic transmission initiated from separate sources for postsynaptic calcium.
Collapse
Affiliation(s)
- H X Chen
- Department of Physiology and Pharmacology, Göteborg University, Sweden
| | | | | | | |
Collapse
|
50
|
Wang D, Maler L. Differential roles of Ca2+/calmodulin-dependent kinases in posttetanic potentiation at input selective glutamatergic pathways. Proc Natl Acad Sci U S A 1998; 95:7133-8. [PMID: 9618551 PMCID: PMC22765 DOI: 10.1073/pnas.95.12.7133] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/1997] [Indexed: 02/07/2023] Open
Abstract
The electrosensory lateral line lobe (ELL) of the electric fish Apteronotus leptorhynchus is a layered medullary region receiving electroreceptor input that terminates on basal dendrites of interneurons and projection (pyramidal) cells. The molecular layer of the ELL contains two distinct glutamatergic feedback pathways that terminate on the proximal (ventral molecular layer, VML) and distal (dorsal molecular layer) apical dendrites of pyramidal cells. Western blot analysis with an antibody directed against mammalian Ca2+/calmodulin-dependent kinase 2, alpha subunit (CaMK2alpha) recognized a protein of identical size in the brain of A. leptorhynchus. Immunohistochemistry demonstrated that CaMK2 alpha expression in the ELL was restricted to fibers and terminals in the VML. Posttetanic potentiation (PTP) could be readily elicited in pyramidal cells by stimulation of either VML or DML in brain slices of the ELL. PTP in the VML was blocked by extracellular application of a CaMK2 antagonist (KN62) while intracellular application of KN62 or a CaMK2 inhibitory peptide had no effect, consistent with the presynaptic localization of CaMK2 alpha in VML. PTP in the dorsal molecular layer was not affected by extracellular application of KN62. Anti-Hebbian plasticity has also been demonstrated in the VML, but was not affected by KN62. These results demonstrate that, while PTP can occur independent of CaMK2, it is, in some synapses, dependent on this kinase.
Collapse
Affiliation(s)
- D Wang
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | | |
Collapse
|