159 results on '"Clive R. Bramham"'
Search Results
2. Generation of an enhancer-driven gene expression viral tool specific to dentate granule cell-types through direct hippocampal injection
- Author
-
Maria Letizia Potenza, Stefan Blankvoort, Miguel M. Carvalho, Joachim S. Grimstvedt, Valentina Di Maria, Kristian Moan, Rajeevkumar Raveendran Nair, Marcus S. Flatset, Qiangwei Zhang, Laurent F. Thomas, Francois P. Pauzin, Rodolfo Da Silva Mazzarini Baldinotti, Giulia Quattrocolo, Clive R. Bramham, Pål Sætrom, Menno P. Witter, and Clifford G. Kentros
- Subjects
rAAVs ,dentate gyrus ,genetic tool ,chromatin immunoprecipitation ,stereotaxic injection ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Accurate investigations of neural circuitry require specific genetic access to individual circuit elements, i.e., the myriad neuronal cell-types in the brain. However, native promoters cannot achieve this because while most genes are expressed in the brain, few are expressed in a single neuronal cell-type. We recently used enhancers, the subcomponents of the transcriptional apparatus which tell promoters when and where to express, combined with heterologous minimal promoters to increase specificity of transgene expression, an approach we call Enhancer-Driven Gene Expression (EDGE). As we discuss, EDGE is a marked improvement in specificity over native promoters, but still requires careful anatomical analysis to avoid off-target effects. In this study we present a more complete set of genomic markers from the mouse brain and characterize a novel EDGE viral vector capable of specifically driving expression in distinct subtypes of hippocampal neurons, even though it can express in other cell-types elsewhere. The advent of cell-type specific viral tools in wild-type animals provides a powerful strategy for neural circuit investigation and holds promise for studies using animal models for which transgenic tools are not available.
- Published
- 2024
- Full Text
- View/download PDF
3. Structural characterization of two nanobodies targeting the ligand-binding pocket of human Arc
- Author
-
José M. Godoy Muñoz, Lasse Neset, Sigurbjörn Markússon, Sarah Weber, Oda C. Krokengen, Aleksi Sutinen, Eleni Christakou, Andrea J. Lopez, Clive R. Bramham, and Petri Kursula
- Subjects
Medicine ,Science - Published
- 2024
4. Editorial: Synaptopathies: from bench to bedside
- Author
-
Clive R. Bramham, Volkmar Lessmann, Anthony J. Hannan, Changhe Wang, Alberto Catanese, Tobias Maria Boeckers, and Hongyu Zhang
- Subjects
synapse ,synaptopathies ,neurodegenerative diseases ,neurodevelopmental disorders ,psychiatric disorders ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Published
- 2023
- Full Text
- View/download PDF
5. Visualizing Arc protein dynamics and localization in the mammalian brain using AAV-mediated in situ gene labeling
- Author
-
Martino Avallone, Joaquín Pardo, Tadiwos F. Mergiya, Jana Rájová, Atte Räsänen, Marcus Davidsson, Malin Åkerblom, Luis Quintino, Darshan Kumar, Clive R. Bramham, and Tomas Björklund
- Subjects
retrotransposon ,synaptic plasticity ,AAV ,PLA ,HITI ,cell–cell transfer ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
The activity-regulated cytoskeleton-associated (Arc) protein is essential for synaptic plasticity and memory formation. The Arc gene, which contains remnants of a structural GAG retrotransposon sequence, produces a protein that self-assembles into capsid-like structures harboring Arc mRNA. Arc capsids, released from neurons, have been proposed as a novel intercellular mechanism for mRNA transmission. Nevertheless, evidence for intercellular transport of Arc in the mammalian brain is still lacking. To enable the tracking of Arc molecules from individual neurons in vivo, we devised an adeno-associated virus (AAV) mediated approach to tag the N-terminal of the mouse Arc protein with a fluorescent reporter using CRISPR/Cas9 homologous independent targeted integration (HITI). We show that a sequence coding for mCherry can successfully be knocked in at the 5′ end of the Arc open reading frame. While nine spCas9 gene editing sites surround the Arc start codon, the accuracy of the editing was highly sequence-dependent, with only a single target resulting in an in-frame reporter integration. When inducing long-term potentiation (LTP) in the hippocampus, we observed an increase of Arc protein highly correlated with an increase in fluorescent intensity and the number of mCherry-positive cells. By proximity ligation assay (PLA), we demonstrated that the mCherry-Arc fusion protein retains the Arc function by interacting with the transmembrane protein stargazin in postsynaptic spines. Finally, we recorded mCherry-Arc interaction with presynaptic protein Bassoon in mCherry-negative surrounding neurons at close proximity to mCherry-positive spines of edited neurons. This is the first study to provide support for inter-neuronal in vivo transfer of Arc in the mammalian brain.
- Published
- 2023
- Full Text
- View/download PDF
6. Detection of Arc/Arg3.1 oligomers in rat brain: constitutive and synaptic activity-evoked dimer expression in vivo
- Author
-
Tadiwos F. Mergiya, Jens Edvard Trygstad Gundersen, Tambudzai Kanhema, Grant Brighter, Yuta Ishizuka, and Clive R. Bramham
- Subjects
Activity-regulated cytoskeleton-associated protein (Arc/Arg3.1) ,brain-derived neurotrophic factor (BDNF) ,dentate gyrus ,in situ protein crosslinking ,long-term potentiation (LTP) ,neuronal cell cultures ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
The immediate early gene product activity-regulated cytoskeleton-associated protein (Arc or Arg3.1) is a major regulator of long-term synaptic plasticity with critical roles in postnatal cortical development and memory formation. However, the molecular basis of Arc function is undefined. Arc is a hub protein with interaction partners in the postsynaptic neuronal compartment and nucleus. Previous in vitro biochemical and biophysical analysis of purified recombinant Arc showed formation of low-order oligomers and larger particles including retrovirus-like capsids. Here, we provide evidence for naturally occurring Arc oligomers in the mammalian brain. Using in situ protein crosslinking to trap weak Arc–Arc interactions, we identified in various preparations a prominent Arc immunoreactive band on SDS-PAGE of molecular mass corresponding to a dimer. While putative trimers, tetramers and heavier Arc species were detected, they were of lower abundance. Stimulus-evoked induction of Arc expression and dimer formation was first demonstrated in SH-SY5Y neuroblastoma cells treated with the muscarinic cholinergic agonist, carbachol, and in primary cortical neuronal cultures treated with brain-derived neurotrophic factor (BDNF). In the dentate gyrus (DG) of adult anesthetized rats, induction of long-term potentiation (LTP) by high-frequency stimulation (HFS) of medial perforant synapses or by brief intrahippocampal infusion of BDNF led to a massive increase in Arc dimer expression. Arc immunoprecipitation of crosslinked DG tissue showed enhanced dimer expression during 4 h of LTP maintenance. Mass spectrometric proteomic analysis of immunoprecipitated, gel-excised bands corroborated detection of Arc dimer. Furthermore, Arc dimer was constitutively expressed in naïve cortical, hippocampal and DG tissue, with the lowest levels in the DG. Taken together the results implicate Arc dimer as the predominant low-oligomeric form in mammalian brain, exhibiting regional differences in its constitutive expression and enhanced synaptic activity-evoked expression in LTP.
- Published
- 2023
- Full Text
- View/download PDF
7. eIF4E phosphorylation recruits β-catenin to mRNA cap and promotes Wnt pathway translation in dentate gyrus LTP maintenance
- Author
-
Sudarshan Patil, Kleanthi Chalkiadaki, Tadiwos F. Mergiya, Konstanze Krimbacher, Inês S. Amorim, Shreeram Akerkar, Christos G. Gkogkas, and Clive R. Bramham
- Subjects
Molecular biology ,Cell biology ,Science - Abstract
Summary: The mRNA cap-binding protein, eukaryotic initiation factor 4E (eIF4E), is crucial for translation and regulated by Ser209 phosphorylation. However, the biochemical and physiological role of eIF4E phosphorylation in translational control of long-term synaptic plasticity is unknown. We demonstrate that phospho-ablated Eif4eS209A Knockin mice are profoundly impaired in dentate gyrus LTP maintenance in vivo, whereas basal perforant path-evoked transmission and LTP induction are intact. mRNA cap-pulldown assays show that phosphorylation is required for synaptic activity-induced removal of translational repressors from eIF4E, allowing initiation complex formation. Using ribosome profiling, we identified selective, phospho-eIF4E-dependent translation of the Wnt signaling pathway in LTP. Surprisingly, the canonical Wnt effector, β-catenin, was massively recruited to the eIF4E cap complex following LTP induction in wild-type, but not Eif4eS209A, mice. These results demonstrate a critical role for activity-evoked eIF4E phosphorylation in dentate gyrus LTP maintenance, remodeling of the mRNA cap-binding complex, and specific translation of the Wnt pathway.
- Published
- 2023
- Full Text
- View/download PDF
8. High-affinity anti-Arc nanobodies provide tools for structural and functional studies
- Author
-
Sigurbjörn Markússon, Erik I. Hallin, Helene J. Bustad, Arne Raasakka, Ju Xu, Gopinath Muruganandam, Remy Loris, Aurora Martinez, Clive R. Bramham, and Petri Kursula
- Subjects
Medicine ,Science - Abstract
Activity-regulated cytoskeleton-associated protein (Arc) is a multidomain protein of retroviral origin with a vital role in the regulation of synaptic plasticity and memory formation in mammals. However, the mechanistic and structural basis of Arc function is poorly understood. Arc has an N-terminal domain (NTD) involved in membrane binding and a C-terminal domain (CTD) that binds postsynaptic protein ligands. In addition, the NTD and CTD both function in Arc oligomerisation, including assembly of retrovirus-like capsids involved in intercellular signalling. To obtain new tools for studies on Arc structure and function, we produced and characterised six high-affinity anti-Arc nanobodies (Nb). The CTD of rat and human Arc were both crystallised in ternary complexes with two Nbs. One Nb bound deep into the stargazin-binding pocket of Arc CTD and suggested competitive binding with Arc ligand peptides. The crystallisation of the human Arc CTD in two different conformations, accompanied by SAXS data and molecular dynamics simulations, paints a dynamic picture of the mammalian Arc CTD. The collapsed conformation closely resembles Drosophila Arc in capsids, suggesting that we have trapped a capsid-like conformation of the human Arc CTD. Our data obtained with the help of anti-Arc Nbs suggest that structural dynamics of the CTD and dimerisation of the NTD may promote the formation of capsids. Taken together, the recombinant high-affinity anti-Arc Nbs are versatile tools that can be further developed for studying mammalian Arc structure and function, as well as mechanisms of Arc capsid formation, both in vitro and in vivo. For example, the Nbs could serve as a genetically encoded tools for inhibition of endogenous Arc interactions in the study of neuronal function and plasticity.
- Published
- 2022
9. Editorial: Coordination of mRNA Transport and Translation With Vesicle and Organelle Trafficking and Dynamics
- Author
-
Anni Vedeler and Clive R. Bramham
- Subjects
RNP granule ,extracellular vesicles ,mRNA ,endosomes ,lysosomes ,Biology (General) ,QH301-705.5 - Published
- 2021
- Full Text
- View/download PDF
10. Structural properties and peptide ligand binding of the capsid homology domains of human Arc
- Author
-
Erik I. Hallin, Clive R. Bramham, and Petri Kursula
- Subjects
Crystal structure ,Specificity ,Peptide binding ,Postsynaptic density ,Protein interaction ,Capsid homology ,Biology (General) ,QH301-705.5 ,Biochemistry ,QD415-436 - Abstract
The activity-regulated cytoskeleton-associated protein (Arc) is important for synaptic plasticity and the normal function of the brain. Arc interacts with neuronal postsynaptic proteins, but the mechanistic details of its function have not been fully established. The C-terminal domain of Arc consists of tandem domains, termed the N- and C-lobe. The N-lobe harbours a peptide binding site, able to bind multiple targets. By measuring the affinity of human Arc towards various peptides from stargazin and guanylate kinase-associated protein (GKAP), we have refined its specificity determinants. We found two sites in the GKAP repeat region that bind to Arc and confirmed these interactions by X-ray crystallography. Phosphorylation of the stargazin peptide did not affect binding affinity but caused changes in thermodynamic parameters. Comparison of the crystal structures of three high-resolution human Arc-peptide complexes identifies three conserved C–H…π interactions at the binding cavity, explaining the sequence specificity of short linear motif binding by Arc. We further characterise central residues of the Arc lobe fold, show the effects of peptide binding on protein dynamics, and identify acyl carrier proteins as structures similar to the Arc lobes. We hypothesise that Arc may affect protein-protein interactions and phase separation at the postsynaptic density, affecting protein turnover and re-modelling of the synapse. The present data on Arc structure and ligand binding will help in further deciphering these processes.
- Published
- 2021
- Full Text
- View/download PDF
11. Herpes Simplex Virus Type 1 Neuronal Infection Triggers the Disassembly of Key Structural Components of Dendritic Spines
- Author
-
Francisca Acuña-Hinrichsen, Adriana Covarrubias-Pinto, Yuta Ishizuka, María Francisca Stolzenbach, Carolina Martin, Paula Salazar, Maite A. Castro, Clive R. Bramham, and Carola Otth
- Subjects
Herpes Simplex Virus Type 1 (HSV-1) ,neurodegeneration ,neurotropic virus ,Arc protein ,memory consolidation ,dendritic spines ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Herpes simplex virus type 1 (HSV-1) is a widespread neurotropic virus. Primary infection of HSV-1 in facial epithelium leads to retrograde axonal transport to the central nervous system (CNS) where it establishes latency. Under stressful conditions, the virus reactivates, and new progeny are transported anterogradely to the primary site of infection. During the late stages of neuronal infection, axonal damage can occur, however, the impact of HSV-1 infection on the morphology and functional integrity of neuronal dendrites during the early stages of infection is unknown. We previously demonstrated that acute HSV-1 infection in neuronal cell lines selectively enhances Arc protein expression - a major regulator of long-term synaptic plasticity and memory consolidation, known for being a protein-interaction hub in the postsynaptic dendritic compartment. Thus, HSV-1 induced Arc expression may alter the functionality of infected neurons and negatively impact dendritic spine dynamics. In this study we demonstrated that HSV-1 infection induces structural disassembly and functional deregulation in cultured cortical neurons, an altered glutamate response, Arc accumulation within the somata, and decreased expression of spine scaffolding-like proteins such as PSD-95, Drebrin and CaMKIIβ. However, whether these alterations are specific to the HSV-1 infection mechanism or reflect a secondary neurodegenerative process remains to be determined.
- Published
- 2021
- Full Text
- View/download PDF
12. Bidirectional Dysregulation of AMPA Receptor-Mediated Synaptic Transmission and Plasticity in Brain Disorders
- Author
-
Hongyu Zhang and Clive R. Bramham
- Subjects
AMPA receptor (AMPAR) ,synaptic transmission and plasticity ,AMPAR trafficking ,neurodegenarative diseases ,neuropsychiatric disorders ,neurodevelopmental disorders ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
AMPA receptors (AMPARs) are glutamate-gated ion channels that mediate the majority of fast excitatory synaptic transmission throughout the brain. Changes in the properties and postsynaptic abundance of AMPARs are pivotal mechanisms in synaptic plasticity, such as long-term potentiation (LTP) and long-term depression (LTD) of synaptic transmission. A wide range of neurodegenerative, neurodevelopmental and neuropsychiatric disorders, despite their extremely diverse etiology, pathogenesis and symptoms, exhibit brain region-specific and AMPAR subunit-specific aberrations in synaptic transmission or plasticity. These include abnormally enhanced or reduced AMPAR-mediated synaptic transmission or plasticity. Bidirectional reversal of these changes by targeting AMPAR subunits or trafficking ameliorates drug-seeking behavior, chronic pain, epileptic seizures, or cognitive deficits. This indicates that bidirectional dysregulation of AMPAR-mediated synaptic transmission or plasticity may contribute to the expression of many brain disorders and therefore serve as a therapeutic target. Here, we provide a synopsis of bidirectional AMPAR dysregulation in animal models of brain disorders and review the preclinical evidence on the therapeutic targeting of AMPARs.
- Published
- 2020
- Full Text
- View/download PDF
13. No Escaping the Rat Race: Simulated Night Shift Work Alters the Time-of-Day Variation in BMAL1 Translational Activity in the Prefrontal Cortex
- Author
-
Andrea R. Marti, Sudarshan Patil, Jelena Mrdalj, Peter Meerlo, Silje Skrede, Ståle Pallesen, Torhild T. Pedersen, Clive R. Bramham, and Janne Grønli
- Subjects
circadian rhythms ,sleep deprivation ,cognition ,synaptic plasticity ,protein synthesis ,eIF4E ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Millions of people worldwide work during the night, resulting in disturbed circadian rhythms and sleep loss. This may cause deficits in cognitive functions, impaired alertness and increased risk of errors and accidents. Disturbed circadian rhythmicity resulting from night shift work could impair brain function and cognition through disrupted synthesis of proteins involved in synaptic plasticity and neuronal function. Recently, the circadian transcription factor brain-and-muscle arnt-like protein 1 (BMAL1) has been identified as a promoter of mRNA translation initiation, the most highly regulated step in protein synthesis, through binding to the mRNA “cap”. In this study we investigated the effects of simulated shift work on protein synthesis markers. Male rats (n = 40) were exposed to forced activity, either in their rest phase (simulated night shift work) or in their active phase (simulated day shift work) for 3 days. Following the third work shift, experimental animals and time-matched undisturbed controls were euthanized (rest work at ZT12; active work at ZT0). Tissue lysates from two brain regions (prefrontal cortex, PFC and hippocampus) implicated in cognition and sleep loss, were analyzed with m7GTP (cap) pull-down to examine time-of-day variation and effects of simulated shift work on cap-bound protein translation. The results show time-of-day variation of protein synthesis markers in PFC, with increased protein synthesis at ZT12. In the hippocampus there was little difference between ZT0 and ZT12. Active phase work did not induce statistically significant changes in protein synthesis markers at ZT0 compared to time-matched undisturbed controls. Rest work, however, resulted in distinct brain-region specific changes of protein synthesis markers compared to time-matched controls at ZT12. While no changes were observed in the hippocampus, phosphorylation of cap-bound BMAL1 and its regulator S6 kinase beta-1 (S6K1) was significantly reduced in the PFC, together with significant reduction in the synaptic plasticity associated protein activity-regulatedcytoskeleton-associated protein (Arc). Our results indicate considerable time-of-day and brain-region specific variation in cap-dependent translation initiation. We concludethat simulated night shift work in rats disrupts the pathways regulating the circadian component of the translation of mRNA in the PFC, and that this may partly explain impaired waking function during night shift work.
- Published
- 2017
- Full Text
- View/download PDF
14. Arc Interacts with the Integral Endoplasmic Reticulum Protein, Calnexin
- Author
-
Craig Myrum, Jonathan Soulé, Margarethe Bittins, Kyle Cavagnini, Kevin Goff, Silje K. Ziemek, Maria S. Eriksen, Sudarshan Patil, Adrian Szum, Rajeevkumar R. Nair, and Clive R. Bramham
- Subjects
arc ,calnexin ,endoplasmic reticulum ,proximity ligation assay ,endocytosis ,synaptic plasticity ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Activity-regulated cytoskeleton-associated protein, Arc, is a major regulator of long-term synaptic plasticity and memory formation. Here we reveal a novel interaction partner of Arc, a resident endoplasmic reticulum transmembrane protein, calnexin. We show an interaction between recombinantly-expressed GST-tagged Arc and endogenous calnexin in HEK293, SH-SY5Y neuroblastoma and PC12 cells. The interaction was dependent on the central linker region of the Arc protein that is also required for endocytosis of AMPA-type glutamate receptors. High-resolution proximity-ligation assays (PLAs) demonstrate molecular proximity of endogenous Arc with the cytosolic C-terminus, but not the lumenal N-terminus of calnexin. In hippocampal neuronal cultures treated with brain-derived neurotrophic factor (BDNF), Arc interacted with calnexin in the perinuclear cytoplasm and dendritic shaft. Arc also interacted with C-terminal calnexin in the adult rat dentate gyrus (DG). After induction of long-term potentiation (LTP) in the perforant path projection to the DG of adult anesthetized rats, enhanced interaction between Arc and calnexin was obtained in the dentate granule cell layer (GCL). Although Arc and calnexin are both implicated in the regulation of receptor endocytosis, no modulation of endocytosis was detected in transferrin uptake assays. Previous work showed that Arc interacts with multiple protein partners to regulate synaptic transmission and nuclear signaling. The identification of calnexin as a binding partner further supports the role of Arc as a hub protein and extends the range of Arc function to the endoplasmic reticulum, though the function of the Arc/calnexin interaction remains to be defined.
- Published
- 2017
- Full Text
- View/download PDF
15. GSK3α and GSK3β Phosphorylate Arc and Regulate its Degradation
- Author
-
Agata Gozdz, Oleksii Nikolaienko, Malgorzata Urbanska, Iwona A. Cymerman, Ewa Sitkiewicz, Magdalena Blazejczyk, Michal Dadlez, Clive R. Bramham, and Jacek Jaworski
- Subjects
Arc/Arg3.1 ,GSK3 ,proteasomal degradation ,phosphorylation ,dendritic spines ,neuronal activity ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
The selective and neuronal activity-dependent degradation of synaptic proteins appears to be crucial for long-term synaptic plasticity. One such protein is activity-regulated cytoskeleton-associated protein (Arc), which regulates the synaptic content of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPAR), excitatory synapse strength and dendritic spine morphology. The levels of Arc protein are tightly regulated, and its removal occurs via proteasome-mediated degradation that requires prior ubiquitination. Glycogen synthase kinases α and β (GSK3α, GSKβ; collectively named GSK3α/β) are serine-threonine kinases with abundant expression in the central nervous system. Both GSK3 isozymes are tonically active under basal conditions, but their activity is regulated by intra- and extracellular factors, intimately involved in neuronal activity. Similar to Arc, GSK3α and GSK3β contribute to synaptic plasticity and the structural plasticity of dendritic spines. The present study identified Arc as a GSK3α/β substrate and showed that GSKβ promotes Arc degradation under conditions that induce de novo Arc synthesis. We also found that GSK3α/β inhibition potentiated spine head thinning that was caused by the prolonged stimulation of N-methyl-D-aspartate receptors (NMDAR). Furthermore, overexpression of Arc mutants that were resistant to GSK3β-mediated phosphorylation or ubiquitination resulted in a stronger reduction of dendritic spine width than wildtype Arc overexpression. Thus, GSK3β terminates Arc expression and limits its effect on dendritic spine morphology. Taken together, the results identify GSK3α/β-catalyzed Arc phosphorylation and degradation as a novel mechanism for controlling the duration of Arc expression and function.
- Published
- 2017
- Full Text
- View/download PDF
16. Dynamic Arc SUMOylation and Selective Interaction with F-Actin-Binding Protein Drebrin A in LTP Consolidation In Vivo
- Author
-
Rajeevkumar R. Nair, Sudarshan Patil, Adrian Tiron, Tambudzai Kanhema, Debabrata Panja, Lars Schiro, Kamil Parobczak, Grzegorz Wilczynski, and Clive R. Bramham
- Subjects
actin cytoskeleton ,brain-derived neurotrophic factor (BDNF) ,dentate gyrus ,hippocampus ,immediate early protein ,long-term potentiation (LTP) ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Activity-regulatedcytoskeleton-associated protein (Arc) protein is implicated as a master regulator of long-term forms of synaptic plasticity and memory formation, but the mechanisms controlling Arc protein function are little known. Post-translation modification by small ubiquitin-like modifier (SUMO) proteins has emerged as a major mechanism for regulating protein-protein interactions and function. We first show in cell lines that ectopically expressed Arc undergoes mono-SUMOylation. The covalent addition of a single SUMO1 protein was confirmed by in vitro SUMOylation of immunoprecipitated Arc. To explore regulation of endogenous Arc during synaptic plasticity, we induced long-term potentiation (LTP) in the dentate gyrus of live anesthetized rats. Using coimmunoprecipitation of native proteins, we show that Arc synthesized during the maintenance phase of LTP undergoes dynamic mono-SUMO1-ylation. Levels of unmodified Arc increase in multiple subcellular fractions (cytosol, membrane, nuclear and cytoskeletal), whereas enhanced Arc SUMOylation was specific to the synaptoneurosomal and the cytoskeletal fractions. Dentate gyrus LTP consolidation requires a period of sustained Arc synthesis driven by brain-derived neurotrophic factor (BDNF) signaling. Local infusion of the BDNF scavenger, TrkB-Fc, during LTP maintenance resulted in rapid reversion of LTP, inhibition of Arc synthesis and loss of enhanced Arc SUMO1ylation. Furthermore, coimmunoprecipitation analysis showed that SUMO1-ylated Arc forms a complex with the F-actin-binding protein drebrin A, a major regulator of cytoskeletal dynamics in dendritic spines. Although Arc also interacted with dynamin 2, calcium/calmodulindependentprotein kinase II-beta (CaMKIIβ), and postsynaptic density protein-95 (PSD-95), these complexes lacked SUMOylated Arc. The results support a model in which newly synthesized Arc is SUMOylated and targeted for actin cytoskeletal regulation during in vivo LTP.
- Published
- 2017
- Full Text
- View/download PDF
17. Two-Stage Translational Control of Dentate Gyrus LTP Consolidation Is Mediated by Sustained BDNF-TrkB Signaling to MNK
- Author
-
Debabrata Panja, Justin W. Kenney, Laura D’Andrea, Francesca Zalfa, Anni Vedeler, Karin Wibrand, Rikiro Fukunaga, Claudia Bagni, Christopher G. Proud, and Clive R. Bramham
- Subjects
Biology (General) ,QH301-705.5 - Abstract
BDNF signaling contributes to protein-synthesis-dependent synaptic plasticity, but the dynamics of TrkB signaling and mechanisms of translation have not been defined. Here, we show that long-term potentiation (LTP) consolidation in the dentate gyrus of live rodents requires sustained (hours) BDNF-TrkB signaling. Surprisingly, this sustained activation maintains an otherwise labile signaling pathway from TrkB to MAP-kinase-interacting kinase (MNK). MNK activity promotes eIF4F translation initiation complex formation and protein synthesis in mechanistically distinct early and late stages. In early-stage translation, MNK triggers release of the CYFIP1/FMRP repressor complex from the 5′-mRNA cap. In late-stage translation, MNK regulates the canonical translational repressor 4E-BP2 in a synapse-compartment-specific manner. This late stage is coupled to MNK-dependent enhanced dendritic mRNA translation. We conclude that LTP consolidation in the dentate gyrus is mediated by sustained BDNF signaling to MNK and MNK-dependent regulation of translation in two functionally and mechanistically distinct stages.
- Published
- 2014
- Full Text
- View/download PDF
18. Environmental Control of Adult Neurogenesis: From Hippocampal Homeostasis to Behavior and Disease
- Author
-
Sjoukje D. Kuipers, Clive R. Bramham, Heather A. Cameron, Carlos P. Fitzsimons, Aniko Korosi, and Paul J. Lucassen
- Subjects
Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Published
- 2014
- Full Text
- View/download PDF
19. Social defeat during adolescence and adulthood differentially induce BDNF-regulated immediate early genes
- Author
-
Caroline M. Coppens, Taweeporn eSiripornmongcolchai, Karin eWibrand, Maria eNordheim Alme, Bauke eBuwalda, Sietse F. De Boer, Jaap M. Koolhaas, and Clive R. Bramham
- Subjects
Hippocampus ,adolescence ,rat ,stress ,synaptic plasticity ,Social Defeat ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Stressful life events generally enhance the vulnerability for the development of human psychopathologies such as anxiety disorders and depression. The incidence rates of adult mental disorders steeply rises during adolescence in parallel with a structural and functional reorganization of the neural circuitry underlying stress reactivity. However, the mechanisms underlying susceptibility to stress and manifestation of mental disorders during adolescence are little understood. We hypothesized that heightened sensitivity to stress during adolescence reflects age-dependent differences in the expression of activity-dependent genes involved in synaptic plasticity. Therefore, we compared the effect of social stress during adolescence with social stress in adulthood on the expression of a panel of genes linked to induction of long-term potentiation (LTP) and brain-derived neurotrophic factor (BDNF) signaling. We show that social defeat during adolescence and adulthood differentially regulates expression of the immediate early genes BDNF, Arc, Carp, and Tieg1, as measured by qPCR in tissue lysates from prefrontal cortex, nucleus accumbens, and hippocampus. In the hippocampus, mRNA levels for all four genes were robustly elevated following social defeat in adolescence, whereas none were induced by defeat in adulthood. The relationship to coping style was also examined using adult reactive and proactive coping rats. Gene expression levels of reactive and proactive animals were similar in the prefrontal cortex and hippocampus. However, a trend toward a differential expression of BDNF and Arc mRNA in the nucleus accumbens was detected. BDNF mRNA was increased in the nucleus accumbens of proactive defeated animals, whereas the expression level in reactive defeated animals was comparable to control animals. The results demonstrate striking differences in immediate early gene expression in response to social defeat in adolescent and adult rats.
- Published
- 2011
- Full Text
- View/download PDF
20. Object-Place Recognition Learning Triggers Rapid Induction of Plasticity-Related Immediate Early Genes and Synaptic Proteins in the Rat Dentate Gyrus
- Author
-
Jonathan Soulé, Zsuzsa Penke, Tambudzai Kanhema, Maria Nordheim Alme, Serge Laroche, and Clive R. Bramham
- Subjects
Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Long-term recognition memory requires protein synthesis, but little is known about the coordinate regulation of specific genes. Here, we examined expression of the plasticity-associated immediate early genes (Arc, Zif268, and Narp) in the dentate gyrus following long-term object-place recognition learning in rats. RT-PCR analysis from dentate gyrus tissue collected shortly after training did not reveal learning-specific changes in Arc mRNA expression. In situ hybridization and immunohistochemistry were therefore used to assess possible sparse effects on gene expression. Learning about objects increased the density of granule cells expressing Arc, and to a lesser extent Narp, specifically in the dorsal blade of the dentate gyrus, while Zif268 expression was elevated across both blades. Thus, object-place recognition triggers rapid, blade-specific upregulation of plasticity-associated immediate early genes. Furthermore, Western blot analysis of dentate gyrus homogenates demonstrated concomitant upregulation of three postsynaptic density proteins (Arc, PSD-95, and α-CaMKII) with key roles in long-term synaptic plasticity and long-term memory.
- Published
- 2008
- Full Text
- View/download PDF
21. Chronic Fluoxetine Treatment Induces Brain Region-Specific Upregulation of Genes Associated with BDNF-Induced Long-Term Potentiation
- Author
-
Maria Nordheim Alme, Karin Wibrand, Grethe Dagestad, and Clive R. Bramham
- Subjects
Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Published
- 2007
- Full Text
- View/download PDF
22. eIF4E phosphorylation recruits β-catenin to mRNA cap and selectively promotes Wnt pathway translation in dentate gyrus LTP maintenance in vivo
- Author
-
Sudarshan Patil, Kleanthi Chalkiadaki, Tadiwos Feyissa Mergiya, Konstanze Simbriger, Inês S. Amorim, Shreeram Akerkar, Christos G. Gkogkas, and Clive R. Bramham
- Abstract
SUMMARYThe mRNA cap-binding protein, eukaryotic initiation factor 4E (eIF4E), is crucial for translation and regulated by Ser209 phosphorylation. However, the biochemical and physiological role of eIF4E phosphorylation in translational control of long-term synaptic plasticity is unknown. We demonstrate that phospho-ablated Eif4eS209A knockin mice are profoundly impaired in dentate gyrus LTP maintenance in vivo, while basal perforant path-evoked transmission and LTP induction are intact. mRNA cap-pulldown assays show that phosphorylation is required for synaptic activity-induced removal of translational repressors from eIF4E, allowing initiation complex formation. Using ribosome profiling, we identified selective, phospho-eIF4E-dependent translation of the Wnt signaling pathway in in vivo LTP. Surprisingly, the canonical Wnt effector, β-catenin, was massively recruited to the eIF4E cap complex following LTP induction in wild-type, but not Eif4eS209A, mice. These results demonstrate a critical role for activity-evoked eIF4E phosphorylation in dentate gyrus LTP maintenance, bidirectional remodeling of the mRNA cap-binding complex, and mRNA-specific translational control linked to Wnt pathway.Key highlightsSynaptic activity-induced eIF4E phosphorylation controls DG-LTP maintenance in vivoeIF4E phosphorylation triggers release of translational repressors from cap complexeIF4E phosphorylation recruits β-catenin to cap complexeIF4E phosphorylation selectively enhances translation of Wnt pathway
- Published
- 2022
- Full Text
- View/download PDF
23. Corrigendum: Dynamic expression of long noncoding RNAs and repeat elements in synaptic plasticity
- Author
-
Jesper Lars Viktor Maag, Debabrata Panja, Ida Sporild, Sudarshan Shantinath Patil, Dominik Conrad Kaczorowski, Clive R Bramham, Marcel E Dinger, and Karin Wibrand
- Subjects
Memory, Long-Term ,Transcription, Genetic ,RNA-Seq ,retrotransposons ,Synaptic plasticity (LTP/LTD) ,long-term potentiation (LTP) ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Published
- 2016
- Full Text
- View/download PDF
24. Activation-induced chromatin reorganization in neurons depends on HDAC1 activity
- Author
-
Agnieszka Grabowska, Hanna Sas-Nowosielska, Bartosz Wojtas, Dagmara Holm-Kaczmarek, Elzbieta Januszewicz, Yana Yushkevich, Iwona Czaban, Pawel Trzaskoma, Katarzyna Krawczyk, Bartlomiej Gielniewski, Ana Martin-Gonzalez, Robert Kuba Filipkowski, Krzysztof Hubert Olszynski, Tytus Bernas, Andrzej Antoni Szczepankiewicz, Malgorzata Alicja Sliwinska, Tambudzai Kanhema, Clive R. Bramham, Grzegorz Bokota, Dariusz Plewczynski, Grzegorz Marek Wilczynski, Adriana Magalska, National Science Centre (Poland), Foundation for Polish Science, and European Commission
- Subjects
Neurons ,Transcription, Genetic ,Long-Term Potentiation ,Histone Deacetylase 1 ,Chromatin condensation ,Chromosomes, Mammalian ,Hippocampus ,General Biochemistry, Genetics and Molecular Biology ,Chromatin ,HDAC1 ,Mice, Inbred C57BL ,Nuclear bodies ,Chromatin organization ,Animals ,Neuronal activation ,Epigenetics ,Calcium Signaling ,Rats, Wistar ,Energy Metabolism - Abstract
Spatial chromatin organization is crucial for transcriptional regulation and might be particularly important in neurons since they dramatically change their transcriptome in response to external stimuli. We show that stimulation of neurons causes condensation of large chromatin domains. This phenomenon can be observed in vitro in cultured rat hippocampal neurons as well as in vivo in the amygdala and hippocampal neurons. Activity-induced chromatin condensation is an active, rapid, energy-dependent, and reversible process. It involves calcium-dependent pathways but is independent of active transcription. It is accompanied by the redistribution of posttranslational histone modifications and rearrangements in the spatial organization of chromosome territories. Moreover, it leads to the reorganization of nuclear speckles and active domains located in their proximity. Finally, we find that the histone deacetylase HDAC1 is the key regulator of this process. Our results suggest that HDAC1-dependent chromatin reorganization constitutes an important level of transcriptional regulation in neurons., This work was supported by the National Science Centre grant nos. UMO-2015/18/E/NZ3/00730 (A.M., A.G., H.S.N., E.J. and Y.Y.), 2014/15/N/NZ2/00379 and 2017/24/T/NZ2/00307 (P.T.), 2019/35/O/ST6/02484 (D.P. and G.B.), and 2014/14/M/NZ4/00561 (K.H.O. and R.K.F.). B.W. and B.G. were supported by the Foundation for Polish Science TEAM-TECH Core Facility project “NGS platform for comprehensive diagnostics and personalized therapy in neuro-oncology,” Foundation for Polish Science co-financed by the European Union under the European Regional Development Fund (TEAM to D.P.). A.M.G. was supported by the H2020-MSCA-COFUND-2014 grant Bio4Med (GA no. 665735).
- Published
- 2022
25. Development and Validation of Arc Nanobodies: New Tools for Probing Arc Dynamics and Function
- Author
-
Yuta Ishizuka, Tadiwos F. Mergiya, Rodolfo Baldinotti, Ju Xu, Erik I. Hallin, Sigurbjörn Markússon, Petri Kursula, and Clive R. Bramham
- Subjects
Mammals ,C-lobe ,Co-immunoprecipitation ,Neuronal Plasticity ,Long-Term Potentiation ,Nerve Tissue Proteins ,Chromobody ,General Medicine ,Single-Domain Antibodies ,Arc ,Biochemistry ,Intrabody ,Rats ,N-lobe ,Cellular and Molecular Neuroscience ,Cytoskeletal Proteins ,Neuroblastoma ,Nanobody ,Animals ,Humans - Abstract
Activity-regulated cytoskeleton-associated (Arc) protein plays key roles in long-term synaptic plasticity, memory, and cognitive flexibility. However, an integral understanding of Arc mechanisms is lacking. Arc is proposed to function as an interaction hub in neuronal dendrites and the nucleus, yet Arc can also form retrovirus-like capsids with proposed roles in intercellular communication. Here, we sought to develop anti-Arc nanobodies (ArcNbs) as new tools for probing Arc dynamics and function. Six ArcNbs representing different clonal lines were selected from immunized alpaca. Immunoblotting with recombinant ArcNbs fused to a small ALFA-epitope tag demonstrated binding to recombinant Arc as well as endogenous Arc from rat cortical tissue. ALFA-ArcNb also provided efficient immunoprecipitation of stimulus-induced Arc after carbachol-treatment of SH-SY5Y neuroblastoma cells and induction of long-term potentiation in the rat dentate gyrus in vivo. Epitope mapping showed that all Nbs recognize the Arc C-terminal region containing the retroviral Gag capsid homology domain, comprised of tandem N-and C-lobes. ArcNbs E5 and H11 selectively bound the N-lobe, which harbors a peptide ligand binding pocket specific to mammals. Four additional ArcNbs bound the region containing the C-lobe and terminal tail. For use as genetically encoded fluorescent intrabodies, we show that ArcNbs fused to mScarlet-I are uniformly expressed, without aggregation, in the cytoplasm and nucleus of HEK293FT cells. Finally, mScarlet-I-ArcNb H11 expressed as intrabody selectively bound the N-lobe and enabled co-immunoprecipitation of full-length intracellular Arc. ArcNbs are versatile tools for live-cell labeling and purification of Arc and analysis of capsid domain specific functions.
- Published
- 2022
26. High-affinity nanobodies as tools for structural and functional studies on mammalian Arc
- Author
-
Sigurbjörn Markússon, Erik I. Hallin, Helene J. Bustad, Arne Raasakka, Ju Xu, Gopinath Muruganandam, Remy Loris, Aurora Martinez, Clive R. Bramham, and Petri Kursula
- Abstract
Activity-regulated cytoskeleton-associated protein (Arc) is a multidomain protein of retroviral origin with a vital role in the regulation of synaptic plasticity and memory formation in mammals. However, the mechanistic and structural basis of Arc function is little understood. Arc has an NTD involved in membrane binding and a CTD which binds postsynaptic protein ligands. In addition, the NTD and CTD both function in Arc oligomerization, including assembly of retrovirus-like capsid involved in intercellular signaling. We produced and characterised six ultra-high-affinity anti-Arc nanobodies (Nb). The CTD of both rat and human Arc could be crystallised in ternary complexes with two Nbs simultaneously bound (H11 and C11). H11 binding deep into the stargazing-binding pocket of Arc CTD suggested competitive binding with Arc ligand peptides, which was confirmed in vitro. This indicates that the H11 Nb could serve as a genetically-encoded tool for inhibition of endogenous Arc N-lobe interactions in study of neuronal function and plasticity. The crystallisation of the human Arc CTD in two different conformations, accompanied by SAXS data and molecular dynamics simulations, paints a dynamic picture of the mammalian Arc CTD. Dynamics were affected by mutations known to inhibit capsid formation, implying a role for Arc CTD dynamics in oligomerisation. Dimerisation of the NTD, together with structural dynamics of the CTD, suggest a mechanism, by which structural dynamics of the CTD may promote capsomer formation, and dimerisation of the NTD links capsomers, facilitating the formation of capsids. The described recombinant ultrahigh-affinity anti-Arc Nbs are versatile tools that can be further developed for studying mammalian Arc structure and function in vitro and in vivo.
- Published
- 2021
- Full Text
- View/download PDF
27. MicroRNA-34a Acutely Regulates Synaptic Efficacy in the Adult Dentate Gyrus In Vivo
- Author
-
Maciej Pajak, T. Ian Simpson, Kine Rønnestad, Clive R. Bramham, Sudarshan Patil, Karin Wibrand, Birgitte Berentsen, and Kevin M Goff
- Subjects
0301 basic medicine ,Long-Term Potentiation ,Neuroscience (miscellaneous) ,Hippocampus ,Biology ,Neurotransmission ,Synaptic Transmission ,Synaptic plasticity ,Rats, Sprague-Dawley ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Synaptic efficacy ,Animals ,Neurons ,Neuronal Plasticity ,Arc (protein) ,microRNA ,Dentate gyrus ,Excitatory Postsynaptic Potentials ,Long-term potentiation ,Rats ,MicroRNAs ,030104 developmental biology ,Neurology ,MicroRNA 34a ,Dentate Gyrus ,Excitatory postsynaptic potential ,Gene expression ,Protein synthesis ,Neuroscience ,miR-34a ,030217 neurology & neurosurgery - Abstract
Activity-dependent synaptic plasticity involves rapid regulation of neuronal protein synthesis on a time-scale of minutes. miRNA function in synaptic plasticity and memory formation has been elucidated by stable experimental manipulation of miRNA expression and activity using transgenic approaches and viral vectors. However, the impact of rapid miRNA modulation on synaptic efficacy is unknown. Here, we examined the effect of acute (12 min), intrahippocampal infusion of a miR-34a antagonist (antimiR) on medial perforant path-evoked synaptic transmission in the dentate gyrus of adult anesthetised rats. AntimiR-34a infusion acutely depressed medial perforant path-evoked field excitatory post-synaptic potentials (fEPSPs). The fEPSP decrease was detected within 9 min of infusion, lasted for hours, and was associated with knockdown of antimiR-34a levels. AntimiR-34a-induced synaptic depression was sequence-specific; no changes were elicited by infusion of scrambled or mismatch control. The rapid modulation suggests that a target, or set of targets, is regulated by miR-34a. Western blot analysis of dentate gyrus lysates revealed enhanced expression of Arc, a known miR-34a target, and four novel predicted targets (Ctip2, PKI-1α, TCF4 and Ube2g1). Remarkably, antimiR-34a had no effect when infused during the maintenance phase of long-term potentiation. We conclude that miR-34a regulates basal synaptic efficacy in the adult dentate gyrus in vivo. To our knowledge, these in vivo findings are the first to demonstrate acute (< 9 min) regulation of synaptic efficacy in the adult brain by a miRNA.
- Published
- 2019
- Full Text
- View/download PDF
28. Sleep and protein synthesis-dependent synaptic plasticity: impacts of sleep loss and stress
- Author
-
Janne eGrønli, Jonathan eSoule, and Clive R Bramham
- Subjects
Brain-Derived Neurotrophic Factor ,Depression ,Gene Expression ,Long-Term Potentiation ,Sleep ,stress ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Sleep has been ascribed a critical role in cognitive functioning. Several lines of evidence implicate sleep in the consolidation of synaptic plasticity and long-term memory. Stress disrupts sleep while impairing synaptic plasticity and cognitive performance. Here, we discuss evidence linking sleep to mechanisms of protein synthesis-dependent synaptic plasticity and synaptic scaling. We then consider how disruption of sleep by acute and chronic stress may impair these mechanisms and degrade sleep function.
- Published
- 2014
- Full Text
- View/download PDF
29. NMDA receptor-dependent regulation of miRNA expression and association with Argonaute during LTP in vivo
- Author
-
Balagopal ePai, Taweeporn eSiripornmongcolchai, Birgitte eBerentsen, Ashraf ePakzad, Christel eVieuille, Ståle ePallesen, Maciej ePajak, T. Ian eSimpson, J Douglas eArmstrong, Karin eWibrand, and Clive R Bramham
- Subjects
Dentate Gyrus ,Gene Expression ,Hippocampus ,RNA-Induced Silencing Complex ,microRNA ,protein synthesis ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
microRNAs (miRNAs) are major regulators of protein synthesis in the brain. A major goal is to identify changes in miRNA expression underlying protein synthesis-dependent forms of synaptic plasticity such as long-term potentiation (LTP). Previous analyses focused on changes in miRNA levels in total lysate samples. Here, we asked whether changes in total miRNA accurately reflect changes in the amount of miRNA bound to Argonaute protein within the miRNA-induced silencing complex (miRISC). Ago2 immunoprecipitation was used to isolate RISC-associated miRNAs following high-frequency stimulation (HFS)-induced LTP in the dentate gyrus of anesthetized rats. Using locked-nucleic acid-based PCR cards for high-throughput screening and independent validation by quantitative TaqMan RT-PCR, we identified differential regulation of Ago2-associated and total miRNA expression. The ratio of Ago2/total miRNA expression was regulated bi-directionally in a miRNA-specific manner and was largely dependent on NMDA receptor activation during LTP induction. The present results identify miRNA association with Ago2 as a potential control point in activity-dependent synaptic plasticity in the adult brain. Finally, novel computational analysis for targets of the Ago2-associated miRNAs identifies 21 pathways that are enriched and differentially targeted by the miRNAs including axon guidance, mTOR, MAPK, Ras and LTP.
- Published
- 2014
- Full Text
- View/download PDF
30. The Changes of the Nuclear Landscape Upon Stimulation of Neuronal Cells are Dependent on the Histone Deacetylase HSAC1
- Author
-
Hanna Sas-Nowosielska, Malgorzata Alicja Sliwinska, Krzysztof H. Olszyński, Pawel Trzaskoma, Iwona Czaban, Dagmara Holm-Kaczmarek, Grzegorz Bokota, Agnieszka Grabowska, Katarzyna Krawczyk, Bartlomiej Gielniewski, Robert K. Filipkowski, Bartosz Wojtas, Ana Martin-Gonzalez, Dariusz Plewczynski, Adriana Magalska, Grzegorz M. Wilczynski, Elzbieta Januszewicz, Tytus Bernas, Clive R. Bramham, Yana Yushkevich, Andrzej Antoni Szczepankiewicz, and Tambudzai Kanhema
- Subjects
Histone ,Prophase ,biology ,Chemistry ,Cellular differentiation ,biology.protein ,Transcriptional regulation ,Stimulation ,Histone deacetylase ,HDAC1 ,Chromatin ,Cell biology - Abstract
Spatial chromatin organization is crucial for transcriptional regulation and might therefore be particularly dynamic in neurons since these terminally differentiated cells dramatically change their transcriptome in response to external stimuli. Here, we show that stimulation of neurons causes condensation of large chromatin domains. We find that this phenomenon is not only induced in rat hippocampal neurons cultured in vitro, but is also present in vivo in amygdala neurons of rats subjected to fear conditioning, and hippocampal neurons of animals subjected to kainate evoked seizures or High-Frequency Stimulation (HFS). The activity-induced chromatin condensation is an active, very rapid, and reversible process, that is independent of transcription and precedes the expression of Immediate Early Genes (IEG). It is accompanied by the redistribution of posttranslational modifications of histones, and rearrangements in the spatial organization of chromosome territories. Moreover, it leads to the reorganization of nuclear speckles and active domains located in their proximity. Finally, we find that neurons depleted of the histone deacetylase HDAC1 fail to condense chromatin upon stimulation, a phenomenon that can be fully reversed by the introduction of human HDAC1. Taken together, our results suggest that the HDAC1-dependent chromatin reorganization might constitute an important level of fine-tuning of transcriptional regulation in stimulated neurons.
- Published
- 2021
- Full Text
- View/download PDF
31. Arc self-association and formation of virus-like capsids are mediated by an N-terminal helical coil motif
- Author
-
Margaret M. Stratton, Erik I. Hallin, Rory O’Connell, Clive R. Bramham, Gopinath Muruganandam, Shreeram Akerkar, James J. Chambers, Yasunori Hayashi, Ian Merski, Petri Kursula, Helene J. Bustad, Remy Loris, Tambudzai Kanhema, Tomohisa Hosokawa, José M. Valpuesta, Marte I. Flydal, Christine Touma, Maria S. Eriksen, Daniela Lascu, Oleksii Nikolaienko, Sverre Grødem, Jorge Cuéllar, Aurora Martinez, Department of Bio-engineering Sciences, Structural Biology Brussels, and Faculty of Sciences and Bioengineering Sciences
- Subjects
0301 basic medicine ,Protein Conformation ,Amino Acid Motifs ,Mutant ,Nerve Tissue Proteins ,Crystallography, X-Ray ,Endocytosis ,Antiparallel (biochemistry) ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Protein Domains ,Animals ,Drosophila Proteins ,Humans ,Protein oligomerization ,Molecular Biology ,Neurons ,Neuronal Plasticity ,Sequence Homology, Amino Acid ,Chemistry ,Cryoelectron Microscopy ,Virion ,RNA ,Cell Biology ,computer.file_format ,Protein Data Bank ,Cytoskeletal Proteins ,030104 developmental biology ,Förster resonance energy transfer ,Capsid ,030220 oncology & carcinogenesis ,Biophysics ,Capsid Proteins ,computer ,Signal Transduction - Abstract
Activity-regulated cytoskeleton-associated protein (Arc) is a protein interaction hub with diverse roles in intracellular neuronal signaling, and important functions in neuronal synaptic plasticity, memory, and postnatal cortical development. Arc has homology to retroviral Gag protein and is capable of self-assembly into virus-like capsids implicated in the intercellular transfer of RNA. However, the molecular basis of Arc self-association and capsid formation is largely unknown. Here, we identified a 28-amino-acid stretch in the mammalian Arc N-terminal (NT) domain that is necessary and sufficient for self-association. Within this region, we identified a 7-residue oligomerization motif, critical for the formation of virus-like capsids. Purified wild-type Arc formed capsids as shown by transmission and cryo-electron microscopy, whereas mutant Arc with disruption of the oligomerization motif formed homogenous dimers. An atomic-resolution crystal structure of the oligomerization region peptide demonstrated an antiparallel coiled-coil interface, strongly supporting NT-NT domain interactions in Arc oligomerization. The NT coil-coil interaction was also validated in live neurons using fluorescence lifetime FRET imaging, and mutation of the oligomerization motif disrupted Arc-facilitated endocytosis. Furthermore, using single-molecule photobleaching, we show that Arc mRNA greatly enhances higher-order oligomerization in a manner dependent on the oligomerization motif. In conclusion, a helical coil in the Arc NT domain supports self-association above the dimer stage, mRNA-induced oligomerization, and formation of virus-like capsids. DATABASE: The coordinates and structure factors for crystallographic analysis of the oligomerization region were deposited at the Protein Data Bank with the entry code 6YTU.
- Published
- 2021
32. Crystal and solution structures reveal oligomerization of individual capsid homology domains of Drosophila Arc
- Author
-
Petri Kursula, Andrew E. Torda, Sigurbjörn Markússon, Lev Böttger, Erik I. Hallin, and Clive R. Bramham
- Subjects
Models, Molecular ,Protein Conformation ,Dimer ,Crystallography, X-Ray ,Biochemistry ,Homology (biology) ,Viral Packaging ,chemistry.chemical_compound ,Protein structure ,0302 clinical medicine ,Animal Cells ,Macromolecular Structure Analysis ,Materials ,Neurons ,0303 health sciences ,Crystallography ,Multidisciplinary ,Arc (protein) ,biology ,Physics ,Monomers ,Condensed Matter Physics ,3. Good health ,Chemistry ,Histone ,Capsid ,Dendritic Structure ,Physical Sciences ,Crystal Structure ,Medicine ,Drosophila ,Cellular Types ,Research Article ,Gene isoform ,Protein Structure ,Science ,Materials Science ,Nerve Tissue Proteins ,Sequence alignment ,Viral Structure ,Microbiology ,DNA-binding protein ,03 medical and health sciences ,Protein Domains ,Virology ,DNA-binding proteins ,Solid State Physics ,Animals ,ddc:610 ,Amino Acid Sequence ,Dimers ,Molecular Biology ,Transcription factor ,030304 developmental biology ,Biology and Life Sciences ,Proteins ,Cell Biology ,Neuronal Dendrites ,Polymer Chemistry ,Solution structure ,Viral Replication ,Cytoskeletal Proteins ,chemistry ,Oligomers ,Cellular Neuroscience ,Synaptic plasticity ,biology.protein ,Biophysics ,Protein Multimerization ,030217 neurology & neurosurgery ,Neuroscience - Abstract
Synaptic plasticity is vital for brain function and memory formation. One of the key proteins in long-term synaptic plasticity and memory is the activity-regulated cytoskeleton-associated protein (Arc). Mammalian Arc forms virus-like capsid structures in a process requiring the N-terminal domain and contains two C-terminal lobes that are structural homologues to retroviral capsids. Drosophila has two isoforms of Arc, dArc1 and dArc2, with low sequence similarity to mammalian Arc, but lacking a large N-terminal domain. Both dArc isoforms are related to the Ty3/gypsy retrotransposon capsid, consisting of N- and C-terminal lobes. Structures of dArc1, as well as capsids formed by both dArc isoforms, have been recently determined. We carried out structural characterization of the four individual dArc lobe domains. As opposed to the corresponding mammalian Arc lobe domains, which are monomeric, the dArc lobes were all oligomeric in solution, indicating a strong propensity for homophilic interactions. A truncated N-lobe from dArc2 formed a domain-swapped dimer in the crystal structure, resulting in a novel dimer interaction that could be relevant for capsid assembly or other dArc functions. This domain-swapped structure resembles the dimeric protein C of flavivirus capsids, as well as the structure of histones dimers, domain-swapped transcription factors, and membrane-interacting BAK domains. The strong oligomerization properties of the isolated dArc lobe domains explain the ability of dArc to form capsids in the absence of any large N-terminal domain, in contrast to the mammalian protein.
- Published
- 2021
- Full Text
- View/download PDF
33. Structural properties and peptide ligand binding of the capsid homology domains of human Arc
- Author
-
Petri Kursula, Erik I. Hallin, and Clive R. Bramham
- Subjects
0301 basic medicine ,QH301-705.5 ,Biophysics ,Peptide ,Peptide binding ,QD415-436 ,Biochemistry ,Homology (biology) ,03 medical and health sciences ,0302 clinical medicine ,Postsynaptic potential ,Capsid homology ,ddc:530 ,Short linear motif ,Biology (General) ,chemistry.chemical_classification ,Arc (protein) ,Chemistry ,Crystal structure ,Protein interaction ,Protein dynamics ,Protein turnover ,Postsynaptic density ,030104 developmental biology ,Capsid ,030220 oncology & carcinogenesis ,Specificity ,Phosphorylation ,Research Article - Abstract
Biochemistry and biophysics reports 26, 100975 (2021). doi:10.1016/j.bbrep.2021.100975, The activity-regulated cytoskeleton-associated protein (Arc) is important for synaptic plasticity and the normal function of the brain. Arc interacts with neuronal postsynaptic proteins, but the mechanistic details of its function have not been fully established. The C-terminal domain of Arc consists of tandem domains, termed the N- and C-lobe. The N-lobe harbours a peptide binding site, able to bind multiple targets. By measuring the affinity of human Arc towards various peptides from stargazin and guanylate kinase-associated protein (GKAP), we have refined its specificity determinants. We found two sites in the GKAP repeat region that bind to Arc and confirmed these interactions by X-ray crystallography. Phosphorylation of the stargazin peptide did not affect binding affinity but caused changes in thermodynamic parameters. Comparison of the crystal structures of three high-resolution human Arc-peptide complexes identifies three conserved C���H����� interactions at the binding cavity, explaining the sequence specificity of short linear motif binding by Arc. We further characterise central residues of the Arc lobe fold, show the effects of peptide binding on protein dynamics, and identify acyl carrier proteins as structures similar to the Arc lobes. We hypothesise that Arc may affect protein-protein interactions and phase separation at the postsynaptic density, affecting protein turnover and re-modelling of the synapse. The present data on Arc structure and ligand binding will help in further deciphering these processes., Published by Elsevier, Amsterdam [u.a.]
- Published
- 2020
- Full Text
- View/download PDF
34. Regulation of Protein Synthesis by eIF4E in the Brain
- Author
-
Christos G. Gkogkas, Clive R. Bramham, Stella Kouloulia, and Kleanthi Chalkiadaki
- Subjects
Chemistry ,EIF4E ,Protein biosynthesis ,Cell biology - Abstract
Regulation of gene expression at the level of mRNA translation is crucial for all the functions our brains carry out. eIF4E binds to the 5′-end of eukaryotic mRNAs and dictates the rate-limiting step of cap-dependent initiation. This chapter reviews the key pathways regulating eIF4E function, but also the less studied and novel mechanisms of eIF4E modulation, linked to synaptic plasticity, learning and memory, and nervous system disorders. Understanding how regulation of protein synthesis by eIF4E affects different aspects of brain function is yet elusive.
- Published
- 2020
- Full Text
- View/download PDF
35. Author response for 'Arc/Arg3.1 function in long‐term synaptic plasticity: emerging mechanisms and unresolved issues'
- Author
-
Clive R. Bramham and Hongyu Zhang
- Subjects
media_common.quotation_subject ,Synaptic plasticity ,Arc arg3 1 ,Biology ,Function (engineering) ,Neuroscience ,Term (time) ,media_common - Published
- 2020
- Full Text
- View/download PDF
36. Herpes Simplex Virus type 1 neuronal infection triggers disassembly of key structural components of dendritic spines
- Author
-
Paula Salazar, Carola Otth, Yuta Ishizuka, Francisca Acuña-Hinrichsen, Maria Francisca Stolzenbach, Maite A. Castro, Carolina Martin, Clive R. Bramham, and Adriana Covarrubias-Pinto
- Subjects
Neurotropic virus ,Arc (protein) ,Dendritic spine ,Neurodegeneration ,Biology ,medicine.disease ,medicine.disease_cause ,Cell biology ,Synapse ,Herpes simplex virus ,nervous system ,Synaptic plasticity ,Axoplasmic transport ,medicine - Abstract
Herpes simplex virus type 1 (HSV-1) is a widespread neurotropic virus. The primary infection in facial epithelium leads to retrograde axonal transport to the central nervous system (CNS) where it establishes latency. Under stressful conditions, the virus reactivates, and new progeny is transported anterogradely to the primary site of infection. In late stages of neuronal infection, axonal damage is known to occur. However, the impact of HSV-1 infection on morphology and functional integrity at earlier stages of infection in neuronal dendrites is unknown. Previously, we demonstrated that acute HSV-1 infection in neuronal cell lines selectively enhances the expression of Arc protein - a major regulator of long-term synaptic plasticity and memory consolidation, known for being a protein-interaction hub in the postsynaptic dendritic compartment. Thus, HSV-1 induced Arc may alter the functionality of the infected neurons having an impact on dendritic spine dynamics. In this study we demonstrated that HSV-1 infection causes structural disassembly and functional deregulation in cultured cortical neurons, through protein homeostasis alteration with intracellular accumulation of Arc, and decreased expression of spine scaffolding-like proteins such as PSD-95, Drebrin and CaMKIIβ. Our findings reveal progressive deleterious effects of HSV-1 infection on excitatory neuronal synapse function and dendritic morphology, supporting the thesis of the infectious origin of neurodegenerative processes. Key words: HSV-1, neurotropic virus, Arc, PSD-95, Drebrin, CaMKIIβ, dendritic spines, neuronal infection, neurodegeneration.
- Published
- 2020
- Full Text
- View/download PDF
37. CREB family transcription factors are major mediators of BDNF transcriptional autoregulation in cortical neurons
- Author
-
Tõnis Timmusk, Alex Sirp, Clive R. Bramham, Jürgen Tuvikene, Sudarshan Patil, and Eli-Eelika Esvald
- Subjects
Male ,0301 basic medicine ,Transcription, Genetic ,MAP Kinase Signaling System ,Nerve Tissue Proteins ,Tropomyosin receptor kinase B ,Biology ,Response Elements ,CREB ,Hippocampus ,Rats, Sprague-Dawley ,03 medical and health sciences ,0302 clinical medicine ,Species Specificity ,Genes, Reporter ,Transduction, Genetic ,Transcription (biology) ,Coactivator ,Genes, Synthetic ,Animals ,Receptor, trkB ,Cyclic AMP Response Element-Binding Protein ,Promoter Regions, Genetic ,Protein Kinase Inhibitors ,Transcription factor ,Cells, Cultured ,Research Articles ,Genes, Dominant ,Cerebral Cortex ,Feedback, Physiological ,Neurons ,Brain-Derived Neurotrophic Factor ,General Neuroscience ,Long-term potentiation ,NFAT ,Recombinant Proteins ,Rats ,Cell biology ,Cytoskeletal Proteins ,Basic-Leucine Zipper Transcription Factors ,030104 developmental biology ,Gene Expression Regulation ,nervous system ,Synaptic plasticity ,biology.protein ,Female ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
BDNF signaling via its transmembrane receptor TrkB has an important role in neuronal survival, differentiation, and synaptic plasticity. Remarkably, BDNF is capable of modulating its own expression levels in neurons, forming a transcriptional positive feedback loop. In the current study, we have investigated this phenomenon in primary cultures of rat cortical neurons using overexpression of dominant-negative forms of several transcription factors, including CREB, ATF2, C/EBP, USF, and NFAT. We show that CREB family transcription factors, together with the coactivator CBP/p300, but not the CRTC family, are the main regulators of ratBDNFgene expression after TrkB signaling. CREB family transcription factors are required for the early induction of all the majorBDNFtranscripts, whereas CREB itself directly binds only toBDNFpromoter IV, is phosphorylated in response to BDNF-TrkB signaling, and activates transcription fromBDNFpromoter IV by recruiting CBP. Our complementary reporter assays withBDNFpromoter constructs indicate that the regulation ofBDNFby CREB family after BDNF-TrkB signaling is generally conserved between rat and human. However, we demonstrate that a nonconserved functional cAMP-responsive element inBDNFpromoter IXa in humans renders the human promoter responsive to BDNF-TrkB-CREB signaling, whereas the rat ortholog is unresponsive. Finally, we show that extensive BDNF transcriptional autoregulation, encompassing all majorBDNFtranscripts, occurs alsoin vivoin the adult rat hippocampus during BDNF-induced LTP. Collectively, these results improve the understanding of the intricate mechanism of BDNF transcriptional autoregulation.SIGNIFICANCE STATEMENTDeeper understanding of stimulus-specific regulation ofBDNFgene expression is essential to precisely adjust BDNF levels that are dysregulated in various neurological disorders. Here, we have elucidated the molecular mechanisms behind TrkB signaling-dependentBDNFmRNA induction and show that CREB family transcription factors are the main regulators ofBDNFgene expression after TrkB signaling. Our results suggest that BDNF-TrkB signaling may induceBDNFgene expression in a distinct manner compared with neuronal activity. Moreover, our data suggest the existence of a stimulus-specific distal enhancer modulatingBDNFgene expression.
- Published
- 2020
- Full Text
- View/download PDF
38. Cognitive function and brain plasticity in a rat model of shift work: role of daily rhythms, sleep and glucocorticoids
- Author
-
Janne Grønli, Jelena Mrdalj, Peter Meerlo, Jonathan P. Wisor, Andrea Rørvik Marti, Øystein Holmelid, Sudarshan Patil, Clive R. Bramham, Torhild Pedersen, and Meerlo lab
- Subjects
Male ,STRESS ,Circadian clock ,lcsh:Medicine ,Morris water navigation task ,NIGHT-SHIFT ,INTERNAL DESYNCHRONIZATION ,Biology ,Article ,Rats, Sprague-Dawley ,Shift work ,Cognition ,Neuroplasticity ,Animals ,TOLERANCE ,Effects of sleep deprivation on cognitive performance ,Circadian rhythm ,Wakefulness ,lcsh:Science ,Glucocorticoids ,Spatial Memory ,Neuronal Plasticity ,Multidisciplinary ,CORTISOL ,MEMORY ,lcsh:R ,Shift Work Schedule ,PERFORMANCE ,Sleep in non-human animals ,Circadian Rhythm ,Rats ,Circadian regulation ,SHORT-TERM ,lcsh:Q ,CIRCADIAN-RHYTHMS ,TRANSLATION ,Sleep ,Neuroscience - Abstract
Many occupations require operations during the night-time when the internal circadian clock promotes sleep, in many cases resulting in impairments in cognitive performance and brain functioning. Here, we use a rat model to attempt to identify the biological mechanisms underlying such impaired performance. Rats were exposed to forced activity, either in their rest-phase (simulating night-shift work; rest work) or in their active-phase (simulating day-shift work; active work). Sleep, wakefulness and body temperature rhythm were monitored throughout. Following three work shifts, spatial memory performance was tested on the Morris Water Maze task. After 4 weeks washout, the work protocol was repeated, and blood and brain tissue collected. Simulated night-shift work impaired spatial memory and altered biochemical markers of cerebral cortical protein synthesis. Measures of daily rhythm strength were blunted, and sleep drive increased. Individual variation in the data suggested differences in shift work tolerance. Hierarchical regression analyses revealed that type of work, changes in daily rhythmicity and changes in sleep drive predict spatial memory performance and expression of brain protein synthesis regulators. Moreover, serum corticosterone levels predicted expression of brain protein synthesis regulators. These findings open new research avenues into the biological mechanisms that underlie individual variation in shift work tolerance.
- Published
- 2020
39. Bidirectional Dysregulation of AMPA Receptor-Mediated Synaptic Transmission and Plasticity in Brain Disorders
- Author
-
Clive R. Bramham and Hongyu Zhang
- Subjects
0301 basic medicine ,Mini Review ,AMPA receptor ,Neurotransmission ,Biology ,lcsh:RC321-571 ,Pathogenesis ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Postsynaptic potential ,medicine ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,neurodegenarative diseases ,Ion channel ,neurodevelopmental disorders ,musculoskeletal, neural, and ocular physiology ,synaptic transmission and plasticity ,AMPAR trafficking ,Chronic pain ,Long-term potentiation ,Cell Biology ,medicine.disease ,neuropsychiatric disorders ,030104 developmental biology ,AMPA receptor (AMPAR) ,nervous system ,Synaptic plasticity ,Neuroscience ,030217 neurology & neurosurgery - Abstract
AMPA receptors (AMPARs) are glutamate-gated ion channels that mediate the majority of fast excitatory synaptic transmission throughout the brain. Changes in the properties and postsynaptic abundance of AMPARs are pivotal mechanisms in synaptic plasticity, such as long-term potentiation (LTP) and long-term depression (LTD) of synaptic transmission. A wide range of neurodegenerative, neurodevelopmental and neuropsychiatric disorders, despite their extremely diverse etiology, pathogenesis and symptoms, exhibit brain region-specific and AMPAR subunit-specific aberrations in synaptic transmission or plasticity. These include abnormally enhanced or reduced AMPAR-mediated synaptic transmission or plasticity. Bidirectional reversal of these changes by targeting AMPAR subunits or trafficking ameliorates drug-seeking behavior, chronic pain, epileptic seizures, or cognitive deficits. This indicates that bidirectional dysregulation of AMPAR-mediated synaptic transmission or plasticity may contribute to the expression of many brain disorders and therefore serve as a therapeutic target. Here, we provide a synopsis of bidirectional AMPAR dysregulation in animal models of brain disorders and review the preclinical evidence on the therapeutic targeting of AMPARs. publishedVersion
- Published
- 2020
40. Correction to: Development and Validation of Arc Nanobodies: New Tools for Probing Arc Dynamics and Function
- Author
-
Yuta Ishizuka, Tadiwos F. Mergiya, Rodolfo Baldinotti, Ju Xu, Erik I. Hallin, Sigurbjörn Markússon, Petri Kursula, and Clive R. Bramham
- Subjects
Cellular and Molecular Neuroscience ,General Medicine ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
41. Structure of monomeric full-length ARC sheds light on molecular flexibility, protein interactions, and functional modalities
- Author
-
Maria S. Eriksen, Oleksii Nikolaienko, Sergei Baryshnikov, Clive R. Bramham, Yasunori Hayashi, Tomohisa Hosokawa, Erik I. Hallin, Petri Kursula, and Sverre Grødem
- Subjects
Male ,Models, Molecular ,0301 basic medicine ,Protein Conformation ,Nerve Tissue Proteins ,Hippocampus ,Biochemistry ,Protein–protein interaction ,Rats, Sprague-Dawley ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Protein structure ,Protein Domains ,Postsynaptic potential ,Fluorescence Resonance Energy Transfer ,Animals ,Humans ,Scattering, Radiation ,Lipid bilayer ,Neurons ,Coiled coil ,Arc (protein) ,Molecular Structure ,Chemistry ,Circular Dichroism ,X-Rays ,Recombinant Proteins ,Rats ,Cytoskeletal Proteins ,030104 developmental biology ,Förster resonance energy transfer ,Biophysics ,CTD ,Postsynaptic signal transduction ,030217 neurology & neurosurgery - Abstract
The activity-regulated cytoskeleton-associated protein (ARC) is critical for long-term synaptic plasticity and memory formation. Acting as a protein interaction hub, ARC regulates diverse signalling events in postsynaptic neurons. A protein interaction site is present in the ARC C-terminal domain (CTD), a bilobar structure homologous to the retroviral Gag capsid domain. We hypothesized that detailed knowledge of the three-dimensional molecular structure of monomeric full-length ARC is crucial to understand its function; therefore, we set out to determine the structure of ARC to understand its various functional modalities. We purified recombinant ARC and analyzed its structure using small-angle X-ray scattering and synchrotron radiation circular dichroism spectroscopy. Monomeric full-length ARC has a compact, closed structure, in which the oppositely charged N-terminal domain (NTD) and CTD are juxtaposed, and the flexible linker between them is not extended. The modeled structure of ARC is supported by intramolecular live-cell Förster resonance energy transfer imaging in rat hippocampal slices. Peptides from several postsynaptic proteins, including stargazin, bind to the N-lobe, but not to the C-lobe, of the bilobar CTD. This interaction does not induce large-scale conformational changes in the CTD or flanking unfolded regions. The ARC NTD contains long helices, predicted to form an anti-parallel coiled coil; binding of ARC to phospholipid membranes requires the NTD. Our data support a role for the ARC NTD in oligomerization as well as lipid membrane binding. The findings have important implications for the structural organization of ARC with respect to distinct functions, such as postsynaptic signal transduction and virus-like capsid formation. Open Practices Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/.
- Published
- 2018
- Full Text
- View/download PDF
42. Arc protein: a flexible hub for synaptic plasticity and cognition
- Author
-
Clive R. Bramham, Maria S. Eriksen, Oleksii Nikolaienko, and Sudarshan Patil
- Subjects
0301 basic medicine ,Memory, Long-Term ,Nonsynaptic plasticity ,Nerve Tissue Proteins ,Biology ,Bioinformatics ,Synaptic Transmission ,Immediate early protein ,Mice ,03 medical and health sciences ,Cognition ,0302 clinical medicine ,Homeostatic plasticity ,Metaplasticity ,Animals ,Humans ,Neuronal Plasticity ,Arc (protein) ,Synaptic scaling ,Brain ,Long-term potentiation ,Cell Biology ,Endocytosis ,Rats ,Cytoskeletal Proteins ,030104 developmental biology ,Receptors, Glutamate ,Synapses ,Synaptic plasticity ,Protein Processing, Post-Translational ,Neuroscience ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Mammalian excitatory synapses express diverse types of synaptic plasticity. A major challenge in neuroscience is to understand how a neuron utilizes different types of plasticity to sculpt brain development, function, and behavior. Neuronal activity-induced expression of the immediate early protein, Arc, is critical for long-term potentiation and depression of synaptic transmission, homeostatic synaptic scaling, and adaptive functions such as long-term memory formation. However, the molecular basis of Arc protein function as a regulator of synaptic plasticity and cognition remains a puzzle. Recent work on the biophysical and structural properties of Arc, its protein-protein interactions and post-translational modifications have shed light on the issue. Here, we present Arc protein as a flexible, multifunctional and interactive hub. Arc interacts with specific effector proteins in neuronal compartments (dendritic spines, nuclear domains) to bidirectionally regulate synaptic strength by distinct molecular mechanisms. Arc stability, subcellular localization, and interactions are dictated by synaptic activity and post-translational modification of Arc. This functional versatility and context-dependent signaling supports a view of Arc as a highly specialized master organizer of long-term synaptic plasticity, critical for information storage and cognition.
- Published
- 2018
- Full Text
- View/download PDF
43. Stimulus-evoked ERK-dependent phosphorylation of activity-regulated cytoskeleton-associated protein (Arc) regulates its neuronal subcellular localization
- Author
-
Sudarshan Patil, Maria S. Eriksen, Oleksii Nikolaienko, Clive R. Bramham, and Haruhiko Bito
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Long-Term Potentiation ,Nerve Tissue Proteins ,Biology ,Hippocampal formation ,Hippocampus ,03 medical and health sciences ,Animals ,Phosphorylation ,Cytoskeleton ,Neurons ,Neuronal Plasticity ,Arc (protein) ,Kinase ,General Neuroscience ,Long-term potentiation ,Subcellular localization ,Molecular biology ,Rats ,Cell biology ,Cytoskeletal Proteins ,030104 developmental biology ,Synapses ,Synaptic plasticity ,Mitogen-Activated Protein Kinases ,Signal Transduction - Abstract
Activity-regulated cytoskeletal-associated protein (Arc) is implicated as a master regulator of long-term synaptic plasticity and memory formation in mammalian brain. Arc acts at synapses and within the nucleus, but the mechanisms controlling Arc localization and function are little known. As Arc transcription and translation are regulated by extracellularsignal-regulated kinase (ERK) signaling, we asked whether Arc protein itself is phosphorylated by ERK. GST-fused Arc of rat origin was able to pull down endogenous ERK2 from rat hippocampal lysates. Using a peptide array, we show that ERK binds a non-canonical docking (D) motif in the C-terminal domain of Arc, and this interaction is abolished by phosphorylation of Tyr309. Activated ERK2 phosphorylated bacterially expressed Arc in vitro at all five predicted sites, as confirmed by phospho-specific protein staining and LC-MS/MS analysis. In neuroblastoma cells expressing epitope tagged-Arc, we demonstrate ERK-dependent phosphorylation of Arc in response to activation of muscarinic cholinergic receptors with carbachol. Using phosphosite-specific antibodies, this stimulus-evoked phosphorylation was shown to occur on Ser206 located within the central hinge region of Arc. In cultured hippocampal neurons expressing phosphomutant Arc under control of the activity-dependent promoter, we show that Ser206 phosphorylation regulates the nuclear:cytosolic localization of Arc. Thus, the neuronal activity-induced phosphomimic exhibits enhanced cytosolic localization relative to phosphodeficient and wild-type Arc. Furthermore, enhanced Ser206 phosphorylation of endogenous Arc was detected in the dentate gyrus cytoskeletal fraction after induction of long-term potentiation (LTP) in live rats. Taken together, this work demonstrates stimulus-evoked ERK-dependent phosphorylation and regulation of Arc protein.
- Published
- 2017
- Full Text
- View/download PDF
44. Antidepressant drugs act by directly binding to TRKB neurotrophin receptors
- Author
-
Eero Castrén, Cassiano R.A.F. Diniz, Stefan Vestring, Timo Petteri Piepponen, Rafael Moliner, Mart Saarma, Anna Steinzeig, Sudarshan Patil, Giray Enkavi, Mykhailo Girych, Clive R. Bramham, Tsvetan Serchov, Katja Kaurinkoski, Plinio C. Casarotto, Madhusmita Pryiadrashini Sahu, Ilpo Vattulainen, Claus Normann, Cecilia A. Brunello, Hanna Antila, Liina Laukkanen, Sari E. Lauri, Cecilia Cannarozzo, Caroline Biojone, Tomasz Róg, Frederike Winkel, Iseline Cardon, Vera Kovaleva, Senem Merve Fred, Neuroscience Center, Materials Physics, Institute of Biotechnology, Faculty of Pharmacy, Department of Physics, Divisions of Faculty of Pharmacy, Division of Pharmacology and Pharmacotherapy, Drug Research Program, Regenerative pharmacology group, Timo Petteri Piepponen / Principal Investigator, Molecular and Integrative Biosciences Research Programme, Synaptic Plasticity and Development, Mart Saarma / Principal Investigator, Tampere University, and Physics
- Subjects
ketamine ,Allosteric regulation ,Tropomyosin receptor kinase B ,Molecular Dynamics Simulation ,114 Physical sciences ,Hippocampus ,Article ,Cell Line ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Protein Domains ,Neurotrophic factors ,Fluoxetine ,Animals ,Humans ,Receptor, trkB ,Binding site ,Receptor ,Visual Cortex ,030304 developmental biology ,0303 health sciences ,antidepressant ,Neuronal Plasticity ,Binding Sites ,molecular dynamic simulation ,biology ,Chemistry ,Brain-Derived Neurotrophic Factor ,musculoskeletal, neural, and ocular physiology ,neurotrophin ,Embryo, Mammalian ,Antidepressive Agents ,Rats ,3. Good health ,Transmembrane domain ,BDNF ,Cholesterol ,nervous system ,plasticity ,Models, Animal ,biology.protein ,1182 Biochemistry, cell and molecular biology ,Antidepressant ,Neuroscience ,030217 neurology & neurosurgery ,Signal Transduction ,Neurotrophin - Abstract
Summary It is unclear how binding of antidepressant drugs to their targets gives rise to the clinical antidepressant effect. We discovered that the transmembrane domain of tyrosine kinase receptor 2 (TRKB), the brain-derived neurotrophic factor (BDNF) receptor that promotes neuronal plasticity and antidepressant responses, has a cholesterol-sensing function that mediates synaptic effects of cholesterol. We then found that both typical and fast-acting antidepressants directly bind to TRKB, thereby facilitating synaptic localization of TRKB and its activation by BDNF. Extensive computational approaches including atomistic molecular dynamics simulations revealed a binding site at the transmembrane region of TRKB dimers. Mutation of the TRKB antidepressant-binding motif impaired cellular, behavioral, and plasticity-promoting responses to antidepressants in vitro and in vivo. We suggest that binding to TRKB and allosteric facilitation of BDNF signaling is the common mechanism for antidepressant action, which may explain why typical antidepressants act slowly and how molecular effects of antidepressants are translated into clinical mood recovery., Graphical Abstract, Highlights • Several antidepressants, including SSRIs and ketamine, directly bind to TRKB • TRKB dimerization at transmembrane region forms a binding pocket for fluoxetine • Antidepressant binding to TRKB facilitates BDNF action and plasticity • Point mutation in TRKB transmembrane region blocks the effects of antidepressants, Direct binding of both typical and fast-acting antidepressants to the BDNF receptor TRKB accounts for cell biological and behavioral actions of antidepressants. This mechanism directly connects antidepressant action to neuronal plasticity and may explain the slow action of typical antidepressants.
- Published
- 2019
- Full Text
- View/download PDF
45. Molecular determinants of Arc oligomerization and formation of virus-like capsids
- Author
-
Clive R. Bramham, Gopinath Muruganandam, Remy Loris, Ian Merski, Marte I. Flydal, Jorge Cuéllar, James J. Chambers, Erik I. Hallin, Yasunori Hayashi, Margaret M. Stratton, Aurora Martinez, Tambudzai Kanhema, José M. Valpuesta, Sverre Grødem, Petri Kursula, Daniela Lascu, Helene J. Bustad, Shreeram Akerkar, Tomohisa Hosokawa, Maria S. Eriksen, Oleksii Nikolaienko, and Rory O’Connell
- Subjects
education.field_of_study ,Arc (protein) ,Capsid ,Chemistry ,Population ,Mutant ,HEK 293 cells ,Biophysics ,RNA ,Alanine scanning ,education ,Green fluorescent protein - Abstract
Expression of activity-regulated cytoskeleton-associated protein (Arc) is critical for long-term synaptic plasticity, memory formation, and cognitive flexibility. The ability of Arc to self-associate and form virus-like capsid structures implies functionally distinct oligomeric states. However, the molecular mechanism of Arc oligomerization is unknown. Here, we identified a 28-amino-acid region necessary and sufficient for Arc oligomerization. This oligomerization region is located within the second coil of a predicted anti-parallel coiled-coil in the N-terminal domain (NTD). Using alanine scanning mutagenesis, we found a 7-amino-acid motif critical for oligomerization and Arc-mediated transferrin endocytosis in HEK cells. Intermolecular fluorescence lifetime imaging in hippocampal neurons confirmed self-association mediated by the motif. To quantify oligomeric size, we performed a single-molecule photobleaching analysis of purified Arc wild-type and mutant. This analysis revealed a critical role for the NTD motif in the formation of higher-order Arc oligomers (30-170 molecules). Moreover, assembly of higher-order wild-type Arc oligomers was significantly enhanced by addition of GFP RNA. Purified wild-type Arc formed virus-like capsids, as visualized by negative-stain EM, and was estimated by light scattering analysis to contain 40-55 Arc units. In contrast, mutant Arc formed a homogenous dimer population as demonstrated by single-molecule TIRF imaging, size-exclusion chromatography with multi-angle light scattering analysis, small-angle X-ray scattering analysis, and single-particle 3D EM reconstruction. Thus, the dimer appears to be the basic building block for assembly. Herein, we show that the NTD motif is essential for higher-order Arc oligomerization, assembly of virus-like capsid particles, and facilitation of oligomerization by exogenous RNA.SIGNIFICANCEArc protein is rapidly expressed in neurons in response to synaptic activity and plays critical roles in synaptic plasticity, postnatal cortical developmental, and memory. Arc has diverse molecular functions, which may be related to distinct oligomeric states of the protein. Arc has homology to retroviral Gag protein and self-assembles into retrovirus-like capsid structures that are capable of intercellular transfer of RNA. Here, we identified a motif in the N-terminal coiled-coil domain of mammalian Arc that mediates higher-order oligomerization and formation of virus-like capsids. The basic building block is the Arc dimer and exogenous RNA facilitates further assembly. The identified molecular determinants of Arc oligomerization will help to elucidate the functional modalities of Arc in the mammalian brain.
- Published
- 2019
- Full Text
- View/download PDF
46. eEF2/eEF2K Pathway in the Mature Dentate Gyrus Determines Neurogenesis Level and Cognition
- Author
-
Iliana Barrera, Sudarshan Patil, Christopher G. Proud, Mohammad Khamaisy, Clive R. Bramham, Elham Taha, Kobi Rosenblum, and Julia Panov
- Subjects
Elongation Factor 2 Kinase ,Male ,0301 basic medicine ,Neurogenesis ,Hippocampus ,Biology ,EEF2 ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,Cognition ,0302 clinical medicine ,Downregulation and upregulation ,Animals ,Phosphorylation ,Mice, Knockout ,Neurons ,Dentate gyrus ,AMPK ,030104 developmental biology ,Dentate Gyrus ,Excitatory postsynaptic potential ,General Agricultural and Biological Sciences ,Neuroscience ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Summary Levels of adult neurogenesis in the dentate gyrus (DG) of the hippocampus are correlated with unique cognitive functions. However, the molecular pathways controlling it are poorly understood. Here, we found that the known physiological ways to enhance neurogenesis converged on the eEF2/eEF2K pathway via AMPK in the DG. Enhancing the elongation phase of mRNA translation in eEF2K-knockout (eEF2K-KO) mice induced the expression of neurogenesis-related proteins in the hippocampus. We thus tested the hypothesis that inducing eEF2K-KO in mature neurons of the DG controls neurogenesis. Indeed, both general eEF2K-KO and targeted KO in DG excitatory mature neurons resulted in enhanced neurogenesis levels and upregulation of neurogenesis-related proteins. Increased neurogenesis was correlated with enhanced performance in DG-dependent learning. Moreover, general and local eEF2K-KO in old mice rejuvenated the DG, paving the way for better mechanistic understanding of how neurogenesis is controlled in the mature DG and possible treatments for incurable aging-associated diseases.
- Published
- 2020
- Full Text
- View/download PDF
47. A simple DMSO-based method for cryopreservation of primary hippocampal and cortical neurons
- Author
-
Yuta Ishizuka and Clive R. Bramham
- Subjects
0301 basic medicine ,Cell viability ,Cell Survival ,Cell ,Hippocampal formation ,Biology ,Stimulus (physiology) ,Hippocampus ,Cryopreservation ,03 medical and health sciences ,0302 clinical medicine ,Gene expression ,medicine ,Animals ,Dimethyl Sulfoxide ,Synapse formation ,Viability assay ,DMSO ,Cells, Cultured ,Neurons ,General Neuroscience ,Primary cultured neuron ,Reproducibility of Results ,Embryo ,Embryonic stem cell ,Synaptic function ,030104 developmental biology ,medicine.anatomical_structure ,nervous system ,Neuronal development ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Background Primary neuronal cultures are widely used to elucidate fundamental aspects of neuronal anatomy, physiology, cell biology, and neuronal dysfunction in animal models of disease. However, preparation of primary neuronal cultures from rodent embryos is labor-intensive, and it is often difficult to produce high-quality cultures consistently in a single laboratory, and to compare results between laboratories. To overcome these issues, cryopreservation can be used to obtain more standardized, high-quality banks of neuronal cultures. New Method In this study, we present a simplified cryopreservation method for rodent primary hippocampal and cortical neurons from embryonic day 18.5 fetuses, using DMSO-containing traditional cell freezing medium. Results Cryopreserved neurons stored for more than 1 year in liquid nitrogen were assessed by cell imaging, as well as biochemical signaling transduction and gene expression in response to pharmacological treatments. Cryopreserved neuronal cultures were comparable to freshly prepared cultures in terms of: (1) neuronal viability, (2) neuronal morphology and maturation, (3) functional synapse formation, (4) stimulus responsiveness. These results indicate that DMSO-cryopreserved neurons are equivalent to freshly prepared neurons both developmentally and functionally. Comparison with Existing Methods Our method is simple and does not require special reagents or equipment. Conclusions Introduction of the cryopreserved neurons as a standard laboratory practice has the potential to increase the robustness and reproducibility of findings between laboratories and reduce the number of animals used in research.
- Published
- 2020
- Full Text
- View/download PDF
48. PKR Inhibition Rescues Memory Deficit and ATF4 Overexpression in ApoE ε4 Human Replacement Mice
- Author
-
Yifat Segev, Tali Rosenberg, Meshi Mints, Clive R. Bramham, Karin Wibrand, Hadile Ounallah-Saad, Iliana Barrera, Orit David, Kobi Rosenblum, Adva Livne, and Ida Sporild
- Subjects
Regulation of gene expression ,medicine.medical_specialty ,biology ,General Neuroscience ,ATF4 ,medicine.disease ,Protein kinase R ,Activating transcription factor 2 ,Endocrinology ,Internal medicine ,medicine ,biology.protein ,Memory impairment ,lipids (amino acids, peptides, and proteins) ,Memory consolidation ,Translation factor ,Alzheimer's disease ,Neuroscience - Abstract
Sporadic Alzheimer's disease (AD) is an incurable neurodegenerative disease with clear pathological hallmarks, brain dysfunction, and unknown etiology. Here, we tested the hypothesis that there is a link between genetic risk factors for AD, cellular metabolic stress, and transcription/translation regulation. In addition, we aimed at reversing the memory impairment observed in a mouse model of sporadic AD. We have previously demonstrated that the most prevalent genetic risk factor for AD, the ApoE4 allele, is correlated with increased phosphorylation of the translation factor eIF2α. In the present study, we tested the possible involvement of additional members of the eIF2α pathway and identified increased mRNA expression of negative transcription factor ATF4 (aka CREB2) both in human and a mouse model expressing the human ApoE4 allele. Furthermore, injection of a PKR inhibitor rescued memory impairment and attenuated ATF4 mRNA increased expression in the ApoE4 mice. The results propose a new mechanism by which ApoE4 affects brain function and further suggest that inhibition of PKR is a way to restore ATF4 overexpression and memory impairment in early stages of sporadic AD.SIGNIFICANCE STATEMENTATF4 mRNA relative quantities are elevated in ApoE4 allele carriers compared with noncarrier controls. This is true also for the ApoE ε4 human replacement mice. ApoE4 mice injected with PKR inhibitor (PKRi) demonstrate a significant reduction in ATF4 expression levels 3 h after one injection of PKRi. Treatment of ApoE4 human replacement mice with the PKRi before learning rescues the memory impairment of the ApoE4 AD model mice. We think that these results propose a new mechanism by which ApoE4 affects brain function and suggest that inhibition of PKR is a way to restore memory impairment in early stages of sporadic AD.
- Published
- 2015
- Full Text
- View/download PDF
49. The RNA-Binding Protein hnRNP K Mediates the Effect of BDNF on Dendritic mRNA Metabolism and Regulates Synaptic NMDA Receptors in Hippocampal Neurons
- Author
-
Birgitte Berentsen, Miranda Mele, Ramiro D. Almeida, Rui Costa, Luís Rodrigues, Ana Rita Santos, Clive R. Bramham, Laura Carreto, Carlos B. Duarte, Maria J. Pinto, Pedro M. Afonso, Pasqualino De Luca, Manuel A. S. Santos, Eduardo Morais, Paulo S. Pinheiro, Diogo Comprido, Sudarshan Patil, Graciano Leal, Ka Wan Li, Molecular and Cellular Neurobiology, AIMMS, Amsterdam Neuroscience - Cellular & Molecular Mechanisms, and Amsterdam Neuroscience - Neurodegeneration
- Subjects
Male ,long-term synaptic potentiation ,Neuronal Excitability ,Tropomyosin receptor kinase B ,Hippocampal formation ,neurotrophins ,Hippocampus ,RNA Transport ,Heterogeneous-Nuclear Ribonucleoprotein K ,Rats, Sprague-Dawley ,0302 clinical medicine ,local translation ,Cells, Cultured ,0303 health sciences ,Chemistry ,General Neuroscience ,Glutamate receptor ,Long-term potentiation ,General Medicine ,New Research ,Cell biology ,NMDA receptor ,Female ,Receptors, N-Methyl-D-Aspartate ,NMDA receptors ,03 medical and health sciences ,Long-term synaptic potentiation ,Animals, Outbred Strains ,Journal Article ,Animals ,Humans ,RNA, Messenger ,Rats, Wistar ,030304 developmental biology ,Brain-Derived Neurotrophic Factor ,Excitatory Postsynaptic Potentials ,Dendrites ,Microarray Analysis ,HEK293 Cells ,BDNF ,nervous system ,6.1 ,Synaptic plasticity ,Synapses ,RNA transport ,Microelectrodes ,030217 neurology & neurosurgery ,Synaptosomes - Abstract
Brain-derived neurotrophic factor (BDNF) is an important mediator of long-term synaptic potentiation (LTP) in the hippocampus. The local effects of BDNF depend on the activation of translation activity, which requires the delivery of transcripts to the synapse. In this work, we found that neuronal activity regulates the dendritic localization of the RNA-binding protein heterogeneous nuclear ribonucleoprotein K (hnRNP K) in cultured rat hippocampal neurons by stimulating BDNF-Trk signaling. Microarray experiments identified a large number of transcripts that are coimmunoprecipitated with hnRNP K, and about 60% of these transcripts are dissociated from the protein upon stimulation of rat hippocampal neurons with BDNF.In vivostudies also showed a role for TrkB signaling in the dissociation of transcripts from hnRNP K upon high-frequency stimulation (HFS) of medial perforant path-granule cell synapses of male rat dentate gyrus (DG). Furthermore, treatment of rat hippocampal synaptoneurosomes with BDNF decreased the coimmunoprecipitation of hnRNP K with mRNAs coding for glutamate receptor subunits, Ca2+- and calmodulin-dependent protein kinase IIβ (CaMKIIβ) and BDNF. Downregulation of hnRNP K impaired the BDNF-induced enhancement of NMDA receptor (NMDAR)-mediated mEPSC, and similar results were obtained upon inhibition of protein synthesis with cycloheximide. The results demonstrate that BDNF regulates specific populations of hnRNP-associated mRNAs in neuronal dendrites and suggests an important role of hnRNP K in BDNF-dependent forms of synaptic plasticity.
- Published
- 2017
- Full Text
- View/download PDF
50. Arc Interacts with the Integral Endoplasmic Reticulum Protein, Calnexin
- Author
-
Kevin M Goff, Sudarshan Patil, Silje K. Ziemek, Margarethe Bittins, Adrian Szum, Jonathan Soule, Maria S. Eriksen, Clive R. Bramham, Craig Myrum, Rajeevkumar Raveendran Nair, and Kyle Cavagnini
- Subjects
0301 basic medicine ,Arc (protein) ,synaptic plasticity ,Endoplasmic reticulum ,Dentate gyrus ,Long-term potentiation ,calnexin ,Biology ,Endocytosis ,arc ,Transmembrane protein ,Cell biology ,lcsh:RC321-571 ,03 medical and health sciences ,endoplasmic reticulum ,Cellular and Molecular Neuroscience ,030104 developmental biology ,Calnexin ,Synaptic plasticity ,endocytosis ,proximity ligation assay ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Original Research ,Neuroscience - Abstract
Activity-regulated cytoskeleton-associated protein, Arc, is a major regulator of long-term synaptic plasticity and memory formation. Here we reveal a novel interaction partner of Arc, a resident endoplasmic reticulum transmembrane protein, calnexin. We show an interaction between recombinantly-expressed GST-tagged Arc and endogenous calnexin in HEK293, SH-SY5Y neuroblastoma and PC12 cells. The interaction was dependent on the central linker region of the Arc protein that is also required for endocytosis of AMPA-type glutamate receptors. High-resolution proximity-ligation assays (PLAs) demonstrate molecular proximity of endogenous Arc with the cytosolic C-terminus, but not the lumenal N-terminus of calnexin. In hippocampal neuronal cultures treated with brain-derived neurotrophic factor (BDNF), Arc interacted with calnexin in the perinuclear cytoplasm and dendritic shaft. Arc also interacted with C-terminal calnexin in the adult rat dentate gyrus (DG). After induction of long-term potentiation (LTP) in the perforant path projection to the DG of adult anesthetized rats, enhanced interaction between Arc and calnexin was obtained in the dentate granule cell layer (GCL). Although Arc and calnexin are both implicated in the regulation of receptor endocytosis, no modulation of endocytosis was detected in transferrin uptake assays. Previous work showed that Arc interacts with multiple protein partners to regulate synaptic transmission and nuclear signaling. The identification of calnexin as a binding partner further supports the role of Arc as a hub protein and extends the range of Arc function to the endoplasmic reticulum, though the function of the Arc/calnexin interaction remains to be defined. publishedVersion
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.