1
|
Zhou L, van Bree N, Boutin L, Ryu J, Moussaud S, Liu M, Otrocka M, Olsson M, Falk A, Wilhelm M. High-throughput neural stem cell-based drug screening identifies S6K1 inhibition as a selective vulnerability in sonic hedgehog-medulloblastoma. Neuro Oncol 2024; 26:1685-1699. [PMID: 38860311 PMCID: PMC11376459 DOI: 10.1093/neuonc/noae104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Medulloblastoma (MB) is one of the most common malignant brain tumors in children. Current treatments have increased overall survival but can lead to devastating side effects and late complications in survivors, emphasizing the need for new, improved targeted therapies that specifically eliminate tumor cells while sparing the normally developing brain. METHODS Here, we used a sonic hedgehog (SHH)-MB model based on a patient-derived neuroepithelial stem cell system for an unbiased high-throughput screen with a library of 172 compounds with known targets. Compounds were evaluated in both healthy neural stem cells (NSCs) and tumor cells derived from the same patient. Based on the difference of cell viability and drug sensitivity score between normal cells and tumor cells, hit compounds were selected and further validated in vitro and in vivo. RESULTS We identified PF4708671 (S6K1 inhibitor) as a potential agent that selectively targets SHH-driven MB tumor cells while sparing NSCs and differentiated neurons. Subsequent validation studies confirmed that PF4708671 inhibited the growth of SHH-MB tumor cells both in vitro and in vivo, and that knockdown of S6K1 resulted in reduced tumor formation. CONCLUSIONS Overall, our results suggest that inhibition of S6K1 specifically affects tumor growth, whereas it has less effect on non-tumor cells. Our data also show that the NES cell platform can be used to identify potentially effective new therapies and targets for SHH-MB.
Collapse
Affiliation(s)
- Leilei Zhou
- Department of Microbiology, Tumor, and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Niek van Bree
- Department of Microbiology, Tumor, and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Lola Boutin
- Department of Microbiology, Tumor, and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Jinhye Ryu
- Department of Microbiology, Tumor, and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Simon Moussaud
- Chemical Biology Consortium Sweden (CBCS), Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Mingzhi Liu
- Department of Microbiology, Tumor, and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Magdalena Otrocka
- Chemical Biology Consortium Sweden (CBCS), Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Magnus Olsson
- Department of Clinical Science, Intervention, and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Anna Falk
- Department of Experimental Medical Science, Lund Stem Cell Center, Lund University, Lund, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Margareta Wilhelm
- Department of Microbiology, Tumor, and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
2
|
MacCallum PE, Cooze JB, Ward J, Moore KA, Blundell J. Evaluating the effects of single, multiple, and delayed systemic rapamycin injections to contextual fear reconsolidation: Implications for the neurobiology of memory and the treatment of PTSD-like re-experiencing. Behav Brain Res 2024; 461:114855. [PMID: 38185381 DOI: 10.1016/j.bbr.2024.114855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/18/2023] [Accepted: 01/03/2024] [Indexed: 01/09/2024]
Abstract
The mechanistic target of rapamycin (mTOR) kinase is known to mediate the formation and persistence of aversive memories. Rapamycin, an mTOR inhibitor, administered around the time of reactivation blocks retrieval-induced mTOR activity and de novo protein synthesis in the brains of rodents, while correspondingly diminishing subsequent fear memory. The goal of the current experiments was to further explore rapamycin's effects on fear memory persistence. First, we examined whether mTOR blockade at different time-points after reactivation attenuates subsequent contextual fear memory. We show that rapamycin treatment 3 or 12 h post-reactivation disrupts memory persistence. Second, we examined whether consecutive days of reactivation paired with rapamycin had additive effects over a single pairing at disrupting a contextual fear memory. We show that additional reactivation-rapamycin pairings exacerbates the reconsolidation impairment. Finally, we examined if impaired reconsolidation of a contextual fear memory from rapamycin treatment had any after-effects on learning and recalling a new fear association. We show that rapamycin-impaired reconsolidation does not affect new learning or recall and protects against fear generalization. Our findings improve our understanding of mTOR- dependent fear memory processes, as well as provide insight into potentially novel treatment options for stress-related psychopathologies such as posttraumatic stress disorder.
Collapse
Affiliation(s)
- Phillip E MacCallum
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Ave, St. John's, Newfoundland and Labrador A1B 3X9, Canada
| | - Jane B Cooze
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Ave, St. John's, Newfoundland and Labrador A1B 3X9, Canada
| | - Joshua Ward
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Ave, St. John's, Newfoundland and Labrador A1B 3X9, Canada
| | - Kelsey Am Moore
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Ave, St. John's, Newfoundland and Labrador A1B 3X9, Canada
| | - Jacqueline Blundell
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Ave, St. John's, Newfoundland and Labrador A1B 3X9, Canada.
| |
Collapse
|
3
|
David J, Mousset M, Trombetti K, Sayasouk B, Neilsen C, Suorsa P, Ruben M, Ruben E, Thiessen J, Pychewicz T, Chu P, Huynh TN. Chronic mild stress leads to anxiety-like behavior and decreased p70 S6K1 activity in the hippocampus of male mice. Physiol Behav 2024; 273:114377. [PMID: 37863347 DOI: 10.1016/j.physbeh.2023.114377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/29/2023] [Accepted: 10/15/2023] [Indexed: 10/22/2023]
Abstract
Major affective disorders are highly prevalent, however, current treatments are limited in their effectiveness due to a lack of understanding of underlying molecular mechanisms. Recent studies have shown that reduced activity of p70 S6 kinase 1 (S6K1), a downstream target of the mechanistic target of rapamycin complex 1 (mTORC1), is linked to anxiety-like behavior in both humans and rodents. The purpose of this study was to investigate the relationship between S6K1 and anxiety-like behavior following chronic mild stress (CMS) and drug-induced inhibition of S6K1. Following CMS, anxiety-like behavior was evaluated using an open field (OF) and elevated plus maze (EPM) in adult male C57/Bl6 mice. After behavior analysis, samples of the hippocampus were harvested for quantification of S6K1, S6 ribosomal protein, glycogen synthase kinase-3 β (GSK3β), and beta tubulin via western blot. Our results demonstrate that CMS mice exhibit anxiety-like behavior in the OF and EPM and reduced activity of S6K1 in the hippocampus (HPC). We measured phosphorylation levels of GSK3β and found that GSK3β phosphorylation was also reduced following CMS compared to control mice. Furthermore, pharmacological inhibition of S6K1 with PF-4708671 in male mice was sufficient to produce anxiety-like behavior in the OF and EPM. These results further support the significant role of S6K1 in the pathogenesis of anxiety and affective disorders.
Collapse
Affiliation(s)
- Jazmine David
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308 United States of America
| | - Marike Mousset
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308 United States of America
| | - Kirby Trombetti
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308 United States of America
| | - Beverly Sayasouk
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308 United States of America
| | - Calvin Neilsen
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308 United States of America
| | - Parker Suorsa
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308 United States of America
| | - Melissa Ruben
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308 United States of America
| | - Elias Ruben
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308 United States of America
| | - Jacob Thiessen
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308 United States of America
| | - Taylor Pychewicz
- Department of Biomedical Sciences, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, United States of America
| | - Ping Chu
- Department of Biochemistry and Molecular Genetics, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, United States of America
| | - Thu N Huynh
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308 United States of America; Department of Biochemistry and Molecular Genetics, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, United States of America.
| |
Collapse
|
4
|
Castillo PE, Jung H, Klann E, Riccio A. Presynaptic Protein Synthesis in Brain Function and Disease. J Neurosci 2023; 43:7483-7488. [PMID: 37940588 PMCID: PMC10634577 DOI: 10.1523/jneurosci.1454-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 11/10/2023] Open
Abstract
Local protein synthesis in mature brain axons regulates the structure and function of presynaptic boutons by adjusting the presynaptic proteome to local demands. This crucial mechanism underlies experience-dependent modifications of brain circuits, and its dysregulation may contribute to brain disorders, such as autism and intellectual disability. Here, we discuss recent advancements in the axonal transcriptome, axonal RNA localization and translation, and the role of presynaptic local translation in synaptic plasticity and memory.
Collapse
Affiliation(s)
- Pablo E Castillo
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Hosung Jung
- Department of Anatomy, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Eric Klann
- Center for Neural Science, New York University, New York, New York 10003
- New York University Neuroscience Institute, New York University Grossman School of Medicine, New York, New York 10016
| | - Antonella Riccio
- UCL Laboratory for Molecular Cell Biology University College London, London, WC1E 6BT, United Kingdom
| |
Collapse
|
5
|
Alapin JM, Mohamed MS, Shrestha P, Khaled HG, Vorabyeva AG, Bowling HL, Oliveira MM, Klann E. Opto4E-BP, an optogenetic tool for inducible, reversible, and cell type-specific inhibition of translation initiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.30.554643. [PMID: 37693507 PMCID: PMC10491233 DOI: 10.1101/2023.08.30.554643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
The protein kinase mechanistic target of rapamycin complex 1 (mTORC1) is one of the primary triggers for initiating cap-dependent translation. Amongst its functions, mTORC1 phosphorylates eIF4E-binding proteins (4E-BPs), which prevents them from binding to eIF4E and thereby enables translation initiation. mTORC1 signaling is required for multiple forms of protein synthesis-dependent synaptic plasticity and various forms of long-term memory (LTM), including associative threat memory. However, the approaches used thus far to target mTORC1 and its effectors, such as pharmacological inhibitors or genetic knockouts, lack fine spatial and temporal control. The development of a conditional and inducible eIF4E knockdown mouse line partially solved the issue of spatial control, but still lacked optimal temporal control to study memory consolidation. Here, we have designed a novel optogenetic tool (Opto4E-BP) for cell type-specific, light-dependent regulation of eIF4E in the brain. We show that light-activation of Opto4E-BP decreases protein synthesis in HEK cells and primary mouse neurons. In situ , light-activation of Opto4E-BP in excitatory neurons decreased protein synthesis in acute amygdala slices. Finally, light activation of Opto4E-BP in principal excitatory neurons in the lateral amygdala (LA) of mice after training blocked the consolidation of LTM. The development of this novel optogenetic tool to modulate eIF4E-dependent translation with spatiotemporal precision will permit future studies to unravel the complex relationship between protein synthesis and the consolidation of LTM.
Collapse
|
6
|
Payne JW, Merza D, Mumby DG. The standard reconsolidation protocol for auditory fear-conditioning does not account for fear to the test context. PLoS One 2023; 18:e0287193. [PMID: 37347748 PMCID: PMC10286967 DOI: 10.1371/journal.pone.0287193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 05/31/2023] [Indexed: 06/24/2023] Open
Abstract
Research on memory reconsolidation has relied heavily on the use of Pavlovian auditory cued-fear conditioning. Here, an auditory cue (CS) is paired with a footshock (US) and the CS is later able to evoke a freezing response when presented alone. Some treatments, when administered to conditioned subjects immediately following a CS-alone (memory reactivation) trial, can attenuate the freezing they display on subsequent CS-alone (test) trials, in the absence of the treatment. This reduction in conditioned freezing is usually taken as evidence that the treatment disrupts post-reactivation reconsolidation of the memory trace representing the pairing of CS and US. We suggest an alternative interpretation that may account, either in whole or in part, for the attenuated freezing. The standard reconsolidation protocol (SRP) for auditory fear-conditioning has a design feature that results in second-order conditioning of fear to the test context, as this context is paired with the fear-evoking CS on the reactivation trial. Since freezing during the CS on the test will reflect the compound influence of contextual-fear and cued-fear, a post-reactivation treatment might attenuate freezing on the test by disrupting consolidation of second-order contextual-fear conditioning, even if it has little or no effect on the stability of the original cued-fear memory. This experiment confirmed that rats tested according to the SRP, in which the reactivation and test trials occur in the same context, freeze more on the test trial than rats that receive the reactivation and test trials in different contexts. This confound could lead to false-positive evidence of disrupted reconsolidation if it is not avoided or minimized, which can be accomplished with a modified protocol.
Collapse
Affiliation(s)
- Jason W. Payne
- Mississauga Department of Psychology, University of Toronto, Mississauga, Ontario, Canada
| | - Devon Merza
- Center for Studies in Behavioural Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
| | - Dave G. Mumby
- Center for Studies in Behavioural Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
| |
Collapse
|
7
|
Goltseker K, Garay P, Bonefas K, Iwase S, Barak S. Alcohol-specific transcriptional dynamics of memory reconsolidation and relapse. Transl Psychiatry 2023; 13:55. [PMID: 36792579 PMCID: PMC9932068 DOI: 10.1038/s41398-023-02352-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 02/17/2023] Open
Abstract
Relapse, a critical issue in alcohol addiction, can be attenuated by disruption of alcohol-associated memories. Memories are thought to temporarily destabilize upon retrieval during the reconsolidation process. Here, we provide evidence for unique transcriptional dynamics underpinning alcohol memory reconsolidation. Using a mouse place-conditioning procedure, we show that alcohol-memory retrieval increases the mRNA expression of immediate-early genes in the dorsal hippocampus and medial prefrontal cortex, and that alcohol seeking is abolished by post-retrieval non-specific inhibition of gene transcription, or by downregulating ARC expression using antisense-oligodeoxynucleotides. However, since retrieval of memories for a natural reward (sucrose) also increased the same immediate-early gene expression, we explored for alcohol-specific transcriptional changes using RNA-sequencing. We revealed a unique transcriptional fingerprint activated by alcohol memories, as the expression of this set of plasticity-related genes was not altered by sucrose-memory retrieval. Our results suggest that alcohol memories may activate two parallel transcription programs: one is involved in memory reconsolidation in general, and another is specifically activated during alcohol-memory processing.
Collapse
Affiliation(s)
- Koral Goltseker
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, 10027, USA
| | - Patricia Garay
- The University of Michigan Neuroscience Graduate Program, Ann Arbor, MI, USA
| | - Katherine Bonefas
- The University of Michigan Neuroscience Graduate Program, Ann Arbor, MI, USA
| | - Shigeki Iwase
- The University of Michigan Neuroscience Graduate Program, Ann Arbor, MI, USA
- Human Genetics Department, The University of Michigan Medical School, University of Michigan, Ann Arbor, MI, 48108, USA
| | - Segev Barak
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, 69978, Israel.
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel.
| |
Collapse
|
8
|
Shi X, von Weltin E, Fitzsimmons E, Do C, Caban Rivera C, Chen C, Liu-Chen LY, Unterwald EM. Reactivation of cocaine contextual memory engages mechanistic target of rapamycin/S6 kinase 1 signaling. Front Pharmacol 2022; 13:976932. [PMID: 36238569 PMCID: PMC9552424 DOI: 10.3389/fphar.2022.976932] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/25/2022] [Indexed: 11/25/2022] Open
Abstract
Mechanistic target of rapamycin (mTOR) C1 and its downstream effectors have been implicated in synaptic plasticity and memory. Our prior work demonstrated that reactivation of cocaine memory engages a signaling pathway consisting of Akt, glycogen synthase kinase-3β (GSK3β), and mTORC1. The present study sought to identify other components of mTORC1 signaling involved in the reconsolidation of cocaine contextual memory, including eukaryotic translation initiation factor 4E (eIF4E)-eIF4G interactions, p70 S6 kinase polypeptide 1 (p70S6K, S6K1) activity, and activity-regulated cytoskeleton (Arc) expression. Cocaine contextual memory was established in adult CD-1 mice using conditioned place preference. After cocaine place preference was established, mice were briefly re-exposed to the cocaine-paired context to reactivate the cocaine memory and brains examined. Western blot analysis showed that phosphorylation of the mTORC1 target, p70S6K, in nucleus accumbens and hippocampus was enhanced 60 min following reactivation of cocaine memories. Inhibition of mTORC1 with systemic administration of rapamycin or inhibition of p70S6K with systemic PF-4708671 after reactivation of cocaine contextual memory abolished the established cocaine place preference. Immunoprecipitation assays showed that reactivation of cocaine memory did not affect eIF4E-eIF4G interactions in nucleus accumbens or hippocampus. Levels of Arc mRNA were significantly elevated 60 and 120 min after cocaine memory reactivation and returned to baseline 24 h later. These findings demonstrate that mTORC1 and p70S6K are required for reconsolidation of cocaine contextual memory.
Collapse
Affiliation(s)
- Xiangdang Shi
- Center for Substance Abuse Research and Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Eva von Weltin
- Center for Substance Abuse Research and Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Emma Fitzsimmons
- Center for Substance Abuse Research and Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Chau Do
- Center for Substance Abuse Research and Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Carolina Caban Rivera
- Center for Substance Abuse Research and Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Chongguang Chen
- Center for Substance Abuse Research and Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Lee-Yuan Liu-Chen
- Center for Substance Abuse Research and Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Ellen M Unterwald
- Center for Substance Abuse Research and Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
9
|
Shrestha P, Klann E. Spatiotemporally resolved protein synthesis as a molecular framework for memory consolidation. Trends Neurosci 2022; 45:297-311. [PMID: 35184897 PMCID: PMC8930706 DOI: 10.1016/j.tins.2022.01.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 01/25/2023]
Abstract
De novo protein synthesis is required for long-term memory consolidation. Dynamic regulation of protein synthesis occurs via a complex interplay of translation factors and modulators. Many components of the protein synthesis machinery have been targeted either pharmacologically or genetically to establish its requirement for memory. The combination of ligand/light-gating and genetic strategies, that is, chemogenetics and optogenetics, has begun to reveal the spatiotemporal resolution of protein synthesis in specific cell types during memory consolidation. This review summarizes current knowledge of the macroscopic and microscopic neural substrates for protein synthesis in memory consolidation. In addition, we highlight future directions for determining the localization and timing of de novo protein synthesis for memory consolidation with tools that permit unprecedented spatiotemporal precision.
Collapse
Affiliation(s)
- Prerana Shrestha
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794, USA.
| | - Eric Klann
- Center for Neural Science, New York University, New York, NY 10012, USA; NYU Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA.
| |
Collapse
|
10
|
Cell-type-specific disruption of PERK-eIF2α signaling in dopaminergic neurons alters motor and cognitive function. Mol Psychiatry 2021; 26:6427-6450. [PMID: 33879865 PMCID: PMC8526653 DOI: 10.1038/s41380-021-01099-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 03/20/2021] [Accepted: 04/01/2021] [Indexed: 02/02/2023]
Abstract
Endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) has been shown to activate the eIF2α kinase PERK to directly regulate translation initiation. Tight control of PERK-eIF2α signaling has been shown to be necessary for normal long-lasting synaptic plasticity and cognitive function, including memory. In contrast, chronic activation of PERK-eIF2α signaling has been shown to contribute to pathophysiology, including memory impairments, associated with multiple neurological diseases, making this pathway an attractive therapeutic target. Herein, using multiple genetic approaches we show that selective deletion of the PERK in mouse midbrain dopaminergic (DA) neurons results in multiple cognitive and motor phenotypes. Conditional expression of phospho-mutant eIF2α in DA neurons recapitulated the phenotypes caused by deletion of PERK, consistent with a causal role of decreased eIF2α phosphorylation for these phenotypes. In addition, deletion of PERK in DA neurons resulted in altered de novo translation, as well as changes in axonal DA release and uptake in the striatum that mirror the pattern of motor changes observed. Taken together, our findings show that proper regulation of PERK-eIF2α signaling in DA neurons is required for normal cognitive and motor function in a non-pathological state, and also provide new insight concerning the onset of neuropsychiatric disorders that accompany UPR failure.
Collapse
|
11
|
Lin R, Learman LN, Na CH, Renuse S, Chen KT, Chen PY, Lee GH, Xiao B, Resnick SM, Troncoso JC, Szumlinski KK, Linden DJ, Park JM, Savonenko A, Pandey A, Worley PF. Persistently Elevated mTOR Complex 1-S6 Kinase 1 Disrupts DARPP-32-Dependent D 1 Dopamine Receptor Signaling and Behaviors. Biol Psychiatry 2021; 89:1058-1072. [PMID: 33353667 PMCID: PMC8076344 DOI: 10.1016/j.biopsych.2020.10.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 10/17/2020] [Accepted: 10/22/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND The serine-threonine kinase mTORC1 (mechanistic target of rapamycin complex 1) is essential for normal cell function but is aberrantly activated in the brain in both genetic-developmental and sporadic diseases and is associated with a spectrum of neuropsychiatric symptoms. The underlying molecular mechanisms of cognitive and neuropsychiatric symptoms remain controversial. METHODS The present study examines behaviors in transgenic models that express Rheb, the most proximal known activator of mTORC1, and profiles striatal phosphoproteomics in a model with persistently elevated mTORC1 signaling. Biochemistry, immunohistochemistry, electrophysiology, and behavior approaches are used to examine the impact of persistently elevated mTORC1 on D1 dopamine receptor (D1R) signaling. The effect of persistently elevated mTORC1 was confirmed using D1-Cre to elevate mTORC1 activity in D1R neurons. RESULTS We report that persistently elevated mTORC1 signaling blocks canonical D1R signaling that is dependent on DARPP-32 (dopamine- and cAMP-regulated neuronal phosphoprotein). The immediate downstream effector of mTORC1, ribosomal S6 kinase 1 (S6K1), phosphorylates and activates DARPP-32. Persistent elevation of mTORC1-S6K1 occludes dynamic D1R signaling downstream of DARPP-32 and blocks multiple D1R responses, including dynamic gene expression, D1R-dependent corticostriatal plasticity, and D1R behavioral responses including sociability. Candidate biomarkers of mTORC1-DARPP-32 occlusion are increased in the brain of human disease subjects in association with elevated mTORC1-S6K1, supporting a role for this mechanism in cognitive disease. CONCLUSIONS The mTORC1-S6K1 intersection with D1R signaling provides a molecular framework to understand the effects of pathological mTORC1 activation on behavioral symptoms in neuropsychiatric disease.
Collapse
Affiliation(s)
- Raozhou Lin
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Lisa N. Learman
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Chan-Hyun Na
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Santosh Renuse
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First ST SW, Rochester, MN 55905, USA.,Center for Individualized Medicine, Mayo Clinic, 200 First ST SW, Rochester, MN 55905, USA
| | - Kevin T. Chen
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Po Yu Chen
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Gum-Hwa Lee
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Bo Xiao
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Susan M. Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD 21224, USA
| | - Juan C. Troncoso
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Karen K. Szumlinski
- Department of Psychological and Brain Sciences and the Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA
| | - David J. Linden
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Joo-Min Park
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Republic of Korea
| | - Alena Savonenko
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First ST SW, Rochester, MN 55905, USA.,Center for Individualized Medicine, Mayo Clinic, 200 First ST SW, Rochester, MN 55905, USA
| | - Paul F. Worley
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Corresponding author. Phone: 410-502-5489
| |
Collapse
|
12
|
MacCallum PE, Blundell J. The mTORC1 inhibitor rapamycin and the mTORC1/2 inhibitor AZD2014 impair the consolidation and persistence of contextual fear memory. Psychopharmacology (Berl) 2020; 237:2795-2808. [PMID: 32601986 DOI: 10.1007/s00213-020-05573-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 05/29/2020] [Indexed: 12/17/2022]
Abstract
RATIONALE The mechanistic target of rapamycin (mTOR) kinase mediates various long-lasting forms of synaptic and behavioural plasticity. However, there is little information concerning the temporal pattern of mTOR activation and susceptibility to pharmacological intervention during consolidation of contextual fear memory. Moreover, the contribution of both mTOR complex 1 and 2 together or the mTOR complex 1 downstream effector p70S6K (S6K1) to consolidation of contextual fear memory is unknown. OBJECTIVE Here, we tested whether different timepoints of vulnerability to rapamycin, a first generation mTOR complex 1 inhibitor, exist for contextual fear memory consolidation and persistence. We also sought to characterize the effects of dually inhibiting mTORC1/2 as well as S6K1 on fear memory formation and persistence. METHODS Rapamycin was injected systemically to mice immediately, 3 h, or 12 h after contextual fear conditioning, and retention was measured at different timepoints thereafter. To determine the effects of a single injection of the dual mTROC1/2 inhibitor AZD2014 after learning on memory consolidation and persistence, a dose-response experiment was carried out. Memory formation and persistence was also assessed in response to the S6K1 inhibitor PF-4708671. RESULTS A single systemic injection of rapamycin immediately or 3 h, but not 12 h, after learning impaired the formation and persistence of contextual fear memory. AZD2014 was found, with limitations, to dose-dependently attenuate memory consolidation and persistence at the highest dose tested (50 mg/kg). In contrast, PF-4708671 had no effect on consolidation or persistence. CONCLUSION Our results indicate the need to further understand the role of mTORC1/2 kinase activity in the molecular mechanisms underlying memory processing and also demonstrate that the effects of mTORC1 inhibition at different timepoints well after learning on memory consolidation and persistence.
Collapse
Affiliation(s)
- Phillip E MacCallum
- Department of Psychology, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Jacqueline Blundell
- Department of Psychology, Memorial University of Newfoundland, St. John's, NL, Canada.
| |
Collapse
|
13
|
Lieberman OJ, Cartocci V, Pigulevskiy I, Molinari M, Carbonell J, Broseta MB, Post MR, Sulzer D, Borgkvist A, Santini E. mTOR Suppresses Macroautophagy During Striatal Postnatal Development and Is Hyperactive in Mouse Models of Autism Spectrum Disorders. Front Cell Neurosci 2020; 14:70. [PMID: 32296308 PMCID: PMC7136750 DOI: 10.3389/fncel.2020.00070] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 03/11/2020] [Indexed: 01/06/2023] Open
Abstract
Macroautophagy (hereafter referred to as autophagy) plays a critical role in neuronal function related to development and degeneration. Here, we investigated whether autophagy is developmentally regulated in the striatum, a brain region implicated in neurodevelopmental disease. We demonstrate that autophagic flux is suppressed during striatal postnatal development, reaching adult levels around postnatal day 28 (P28). We also find that mTOR signaling, a key regulator of autophagy, increases during the same developmental period. We further show that mTOR signaling is responsible for suppressing autophagy, via regulation of Beclin-1 and VPS34 activity. Finally, we discover that autophagy is downregulated during late striatal postnatal development (P28) in mice with in utero exposure to valproic acid (VPA), an established mouse model of autism spectrum disorder (ASD). VPA-exposed mice also display deficits in striatal neurotransmission and social behavior. Correction of hyperactive mTOR signaling in VPA-exposed mice restores social behavior. These results demonstrate that neurons coopt metabolic signaling cascades to developmentally regulate autophagy and provide additional evidence that mTOR-dependent signaling pathways represent pathogenic signaling cascades in ASD mouse models that are active during specific postnatal windows.
Collapse
Affiliation(s)
- Ori J. Lieberman
- Division of Molecular Therapeutics, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
- Division of Movement Disorders, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | | | - Irena Pigulevskiy
- Division of Movement Disorders, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Maya Molinari
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Josep Carbonell
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | | | - Michael R. Post
- Division of Molecular Therapeutics, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - David Sulzer
- Division of Molecular Therapeutics, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Anders Borgkvist
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Emanuela Santini
- Division of Movement Disorders, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
14
|
Venkatasubramani JP, Subramanyam P, Pal R, Reddy BK, Srinivasan DJ, Chattarji S, Iossifov I, Klann E, Bhattacharya A. N-terminal variant Asp14Asn of the human p70 S6 Kinase 1 enhances translational signaling causing different effects in developing and mature neuronal cells. Neurobiol Learn Mem 2020; 171:107203. [PMID: 32147585 DOI: 10.1016/j.nlm.2020.107203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 01/23/2020] [Accepted: 02/29/2020] [Indexed: 01/02/2023]
Abstract
The ribosomal p70 S6 Kinase 1 (S6K1) has been implicated in the etiology of complex neurological diseases including autism, depression and dementia. Though no major gene disruption has been reported in humans in RPS6KB1, single nucleotide variants (SNVs) causing missense mutations have been identified, which have not been assessed for their impact on protein function. These S6K1 mutations have the potential to influence disease progression and treatment response. We mined the Simon Simplex Collection (SSC) and SPARK autism database to find inherited SNVs in S6K1 and characterized the effect of two missense SNVs, Asp14Asn (allele frequency = 0.03282%) and Glu44Gln (allele frequency = 0.0008244%), on S6K1 function in HEK293, human ES cells and primary neurons. Expressing Asp14Asn in HEK293 cells resulted in increased basal phosphorylation of downstream targets of S6K1 and increased de novo translation. This variant also showed blunted response to the specific S6K1 inhibitor, FS-115. In human embryonic cell line Shef4, Asp14Asn enhanced spontaneous neural fate specification in the absence of differentiating growth factors. In addition to enhanced translation, neurons expressing Asp14Asn exhibited impaired dendritic arborization and increased levels of phosphorylated ERK 1/2. Finally, in the SSC families tracked, Asp14Asn segregated with lower IQ scores when found in the autistic individual rather than the unaffected sibling. The Glu44Gln mutation showed a milder, but opposite phenotype in HEK cells as compared to Asp14Asn. Although the Glu44Gln mutation displayed increased neuronal translation, it had no impact on neuronal morphology. Our results provide the first characterization of naturally occurring human S6K1 variants on cognitive phenotype, neuronal morphology and maturation, underscoring again the importance of translation control in neural development and plasticity.
Collapse
Affiliation(s)
- Janani Priya Venkatasubramani
- Centre for Brain Development and Repair, Institute for Stem Cell Science and Regenerative Medicine, GKVK Post, Bellary Road, Bangalore, India
| | - Prakash Subramanyam
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY, USA
| | - Rakhi Pal
- Centre for Brain Development and Repair, Institute for Stem Cell Science and Regenerative Medicine, GKVK Post, Bellary Road, Bangalore, India
| | - Bharath K Reddy
- Centre for Brain Development and Repair, Institute for Stem Cell Science and Regenerative Medicine, GKVK Post, Bellary Road, Bangalore, India
| | - Durga Jeyalakshmi Srinivasan
- Centre for Brain Development and Repair, Institute for Stem Cell Science and Regenerative Medicine, GKVK Post, Bellary Road, Bangalore, India; University of Trans-Disciplinary Health Sciences and Technology, Bangalore, India
| | - Sumantra Chattarji
- Centre for Brain Development and Repair, Institute for Stem Cell Science and Regenerative Medicine, GKVK Post, Bellary Road, Bangalore, India; National Centre for Biological Sciences, Tata Institute of Fundamental Research, GKVK Post, Bellary Road, Bangalore, India
| | - Ivan Iossifov
- Cold Spring Harbor Laboratory, 1 Bungtown Rd., Cold Spring Harbor, New York, NY, USA
| | - Eric Klann
- Center for Neural Science, New York University, 4 Washington Place New York, NY, USA
| | - Aditi Bhattacharya
- Centre for Brain Development and Repair, Institute for Stem Cell Science and Regenerative Medicine, GKVK Post, Bellary Road, Bangalore, India.
| |
Collapse
|
15
|
Lieberman OJ, Cartocci V, Pigulevskiy I, Molinari M, Carbonell J, Broseta MB, Post MR, Sulzer D, Borgkvist A, Santini E. mTOR Suppresses Macroautophagy During Striatal Postnatal Development and Is Hyperactive in Mouse Models of Autism Spectrum Disorders. Front Cell Neurosci 2020; 14:70. [PMID: 32296308 DOI: 10.3389/fncel.2020.00070/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 03/11/2020] [Indexed: 05/20/2023] Open
Abstract
Macroautophagy (hereafter referred to as autophagy) plays a critical role in neuronal function related to development and degeneration. Here, we investigated whether autophagy is developmentally regulated in the striatum, a brain region implicated in neurodevelopmental disease. We demonstrate that autophagic flux is suppressed during striatal postnatal development, reaching adult levels around postnatal day 28 (P28). We also find that mTOR signaling, a key regulator of autophagy, increases during the same developmental period. We further show that mTOR signaling is responsible for suppressing autophagy, via regulation of Beclin-1 and VPS34 activity. Finally, we discover that autophagy is downregulated during late striatal postnatal development (P28) in mice with in utero exposure to valproic acid (VPA), an established mouse model of autism spectrum disorder (ASD). VPA-exposed mice also display deficits in striatal neurotransmission and social behavior. Correction of hyperactive mTOR signaling in VPA-exposed mice restores social behavior. These results demonstrate that neurons coopt metabolic signaling cascades to developmentally regulate autophagy and provide additional evidence that mTOR-dependent signaling pathways represent pathogenic signaling cascades in ASD mouse models that are active during specific postnatal windows.
Collapse
Affiliation(s)
- Ori J Lieberman
- Division of Molecular Therapeutics, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
- Division of Movement Disorders, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | | | - Irena Pigulevskiy
- Division of Movement Disorders, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Maya Molinari
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Josep Carbonell
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | | | - Michael R Post
- Division of Molecular Therapeutics, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - David Sulzer
- Division of Molecular Therapeutics, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Anders Borgkvist
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Emanuela Santini
- Division of Movement Disorders, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
16
|
Kats IR, Klann E. Translating from cancer to the brain: regulation of protein synthesis by eIF4F. ACTA ACUST UNITED AC 2019; 26:332-342. [PMID: 31416906 PMCID: PMC6699409 DOI: 10.1101/lm.050047.119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 07/03/2019] [Indexed: 12/27/2022]
Abstract
Formation of eukaryotic initiation factor 4F (eIF4F) is widely considered to be the rate-limiting step in cap-dependent translation initiation. Components of eIF4F are often up-regulated in various cancers, and much work has been done to elucidate the role of each of the translation initiation factors in cancer cell growth and survival. In fact, many of the basic mechanisms describing how eIF4F is assembled and how it functions to regulate translation initiation were first investigated in cancer cell lines. These same eIF4F translational control pathways also are relevant for neuronal signaling that underlies long-lasting synaptic plasticity and memory, and in neurological diseases where eIF4F and its upstream regulators are dysregulated. Although eIF4F is important in cancer and for brain function, there is not always a clear path to use the results of studies performed in cancer models to inform one of the roles that the same translation factors have in neuronal signaling. Issues arise when extrapolating from cell lines to tissue, and differences are likely to exist in how eIF4F and its upstream regulatory pathways are expressed in the diverse neuronal subtypes found in the brain. This review focuses on summarizing the role of eIF4F and its accessory proteins in cancer, and how this information has been utilized to investigate neuronal signaling, synaptic function, and animal behavior. Certain aspects of eIF4F regulation are consistent across cancer and neuroscience, whereas some results are more complicated to interpret, likely due to differences in the complexity of the brain, its billions of neurons and synapses, and its diverse cell types.
Collapse
Affiliation(s)
- Ilona R Kats
- Sackler Graduate Program, New York University School of Medicine, New York, New York 10016, USA.,Center for Neural Science, New York University, New York, New York 10003, USA
| | - Eric Klann
- Center for Neural Science, New York University, New York, New York 10003, USA.,NYU Neuroscience Institute, New York University School of Medicine, New York, New York 10016, USA
| |
Collapse
|
17
|
Inositol polyphosphate multikinase mediates extinction of fear memory. Proc Natl Acad Sci U S A 2019; 116:2707-2712. [PMID: 30692248 DOI: 10.1073/pnas.1812771116] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Inositol polyphosphate multikinase (IPMK), the key enzyme for the biosynthesis of higher inositol polyphosphates and phosphatidylinositol 3,4,5-trisphosphate, also acts as a versatile signaling player in regulating tissue growth and metabolism. To elucidate neurobehavioral functions of IPMK, we generated mice in which IPMK was deleted from the excitatory neurons of the postnatal forebrain. These mice showed no deficits in either novel object recognition or spatial memory. IPMK conditional knockout mice formed cued fear memory normally but displayed enhanced fear extinction. Signaling analyses revealed dysregulated expression of neural genes accompanied by selective activation of the mechanistic target of rapamycin (mTOR) regulatory enzyme p85 S6 kinase 1 (S6K1) in the amygdala following fear extinction. The IPMK mutants also manifested facilitated hippocampal long-term potentiation. These findings establish a signaling action of IPMK that mediates fear extinction.
Collapse
|
18
|
Kataoka T, Fuchikami M, Nojima S, Nagashima N, Araki M, Omura J, Miyagi T, Okamoto Y, Morinobu S. Combined brain-derived neurotrophic factor with extinction training alleviate impaired fear extinction in an animal model of post-traumatic stress disorder. GENES BRAIN AND BEHAVIOR 2018; 18:e12520. [PMID: 30246290 DOI: 10.1111/gbb.12520] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 09/18/2018] [Accepted: 09/18/2018] [Indexed: 01/08/2023]
Abstract
Impaired fear memory extinction (Ext) is one of the hallmark symptoms of post-traumatic stress disorder (PTSD). However, since the precise mechanism of impaired Ext remains unknown, effective interventions have not yet been established. Recently, hippocampal-prefrontal brain-derived neurotrophic factor (BDNF) activity was shown to be crucial for Ext in naïve rats. We therefore examined whether decreased hippocampal-prefrontal BDNF activity is also involved in the Ext of rats subjected to a single prolonged stress (SPS) as a model of PTSD. BDNF levels were measured by enzyme-linked immunosorbent assay (ELISA), and phosphorylation of TrkB was measured by immunohistochemistry in the hippocampus and medial prefrontal cortex (mPFC) of SPS rats. We also examined whether BDNF infusion into the ventral mPFC or hippocampus alleviated the impaired Ext of SPS rats in the contextual fear conditioning paradigm. SPS significantly decreased the levels of BDNF in both the hippocampus and mPFC and TrkB phosphorylation in the ventral mPFC. Infusion of BDNF 24 hours after conditioning in the infralimbic cortex (ILC), but not the prelimbic cortex (PLC) nor hippocampus, alleviated the impairment of Ext. Since amelioration of impaired Ext by BDNF infusion did not occur without extinction training, it seems the two interventions must occur consecutively to alleviate impaired Ext. Additionally, BDNF infusion markedly increased TrkB phosphorylation in the ILC of SPS rats. These findings suggest that decreased BDNF signal transduction might be involved in the impaired Ext of SPS rats, and that activation of the BDNF-TrkB signal might be a novel therapeutic strategy for the impaired Ext by stress.
Collapse
Affiliation(s)
- Tsutomu Kataoka
- Department of Psychiatry and Neurosciences, Division of Frontier Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Manabu Fuchikami
- Department of Psychiatry and Neurosciences, Division of Frontier Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Shinji Nojima
- Department of Psychiatry and Neurosciences, Division of Frontier Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Nobuyuki Nagashima
- Department of Psychiatry and Neurosciences, Division of Frontier Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Motoaki Araki
- Department of Psychiatry and Neurosciences, Division of Frontier Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Jun Omura
- Department of Psychiatry and Neurosciences, Division of Frontier Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Tatsuhiro Miyagi
- Department of Psychiatry and Neurosciences, Division of Frontier Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Yasumasa Okamoto
- Department of Psychiatry and Neurosciences, Division of Frontier Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Shigeru Morinobu
- Department of Occupational Therapy, School of Health Science and Social Welfare, Kibi International University, Takahashi, Japan
| |
Collapse
|
19
|
Wang SH. Novelty enhances memory persistence and remediates propranolol-induced deficit via reconsolidation. Neuropharmacology 2018; 141:42-54. [PMID: 30125560 PMCID: PMC6178872 DOI: 10.1016/j.neuropharm.2018.08.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 08/02/2018] [Accepted: 08/16/2018] [Indexed: 11/23/2022]
Abstract
Memory reactivation has been shown to open a time window for memory modulation. The majority of the methodological or pharmacological approaches target disruption of reconsolidation to weaken aversive memories. However, methods to improve appetitive memory persistence through reconsolidation or to reverse drug-induced reconsolidation impairment are limited. To improve memory persistence, previous studies show that a novel event, introduced around the time of memory encoding, enables the persistence of an otherwise decayed memory. This is mainly through a memory consolidation process. The current study first investigated if a novel event introduced during memory reactivation improves memory persistence through reconsolidation. Using a rodent appetitive spatial paradigm, similar to the human everyday experience of recalling where an item is located, a novel event around memory reactivation facilitated the persistence of spatial memory. This facilitation did not occur when the novel event was omitted and the protein synthesis-dependent reconsolidation was not affected by zif268 anti-sense in the dorsal hippocampus. Furthermore, beta-adrenergic antagonists, propranolol, impaired reconsolidation of appetitive spatial memory and contextual fear conditioning. A novel event after memory reactivation could reverse this impairment due to propranolol. Together, this study provides methods and confirmation for improving memory persistence during memory reactivation and reconsolidation. A novel event can reverse memory impairment caused by interfering reconsolidation with a noradrenergic β-blocker. Immediate-early gene, zif268, is not required for protein synthesis-dependent reconsolidation of appetitive spatial memory. A novel event can reverse the memory impairment caused by blocking reconsolidation with the noradrenergic beta-blocker propranolol.
Collapse
Affiliation(s)
- Szu-Han Wang
- Centre for Clinical Brain Sciences, Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK.
| |
Collapse
|
20
|
Activation of a novel p70 S6 kinase 1-dependent intracellular cascade in the basolateral nucleus of the amygdala is required for the acquisition of extinction memory. Mol Psychiatry 2018; 23:1394-1401. [PMID: 28461701 PMCID: PMC5668214 DOI: 10.1038/mp.2017.99] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 03/21/2017] [Accepted: 03/27/2017] [Indexed: 12/13/2022]
Abstract
Repeated presentations of a previously conditioned stimulus lead to a new form of learning known as extinction, which temporarily alters the response to the original stimulus. Previous studies have shown that the consolidation of extinction memory requires de novo protein synthesis. However, the role of specific nodes of translational control in extinction is unknown. Using auditory threat conditioning in mice, we investigated the role of mechanistic target of rapamycin complex 1 (mTORC1) and its effector p70 S6 kinase 1 (S6K1) in the extinction of auditory threat conditioning. We found that rapamycin attenuated the consolidation of extinction memory. In contrast, genetic deletion and pharmacological inhibition of S6K1, a downstream effector of mTORC1, blocked within-session extinction, indicating a role for S6K1 independent of protein synthesis. Indeed, the activation of S6K1 during extinction required extracellular signal-regulated kinase (ERK) activation in the basolateral nucleus of the amygdala (BLA) and was necessary for increased phosphorylation of the GluA1 (Thr840) subunit of the AMPA receptor following extinction training. Mice exposed to brief uncontrollable stress showed impaired within-session extinction as well as a downregulation of ERK and S6K1 signaling in the amygdala. Finally, using fiber photometry we were able to record calcium signals in vivo, and we found that inhibition of S6K1 reduces extinction-induced changes in neuronal activity of the BLA. These results implicate a novel ERK-S6K1-GluA1 signaling cascade critically involved in extinction.
Collapse
|
21
|
Santini E, Huynh TN, Longo F, Koo SY, Mojica E, D'Andrea L, Bagni C, Klann E. Reducing eIF4E-eIF4G interactions restores the balance between protein synthesis and actin dynamics in fragile X syndrome model mice. Sci Signal 2017; 10:10/504/eaan0665. [PMID: 29114037 DOI: 10.1126/scisignal.aan0665] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Fragile X syndrome (FXS) is the most common form of inherited intellectual disability and autism spectrum disorder. FXS is caused by silencing of the FMR1 gene, which encodes fragile X mental retardation protein (FMRP), an mRNA-binding protein that represses the translation of its target mRNAs. One mechanism by which FMRP represses translation is through its association with cytoplasmic FMRP-interacting protein 1 (CYFIP1), which subsequently sequesters and inhibits eukaryotic initiation factor 4E (eIF4E). CYFIP1 shuttles between the FMRP-eIF4E complex and the Rac1-Wave regulatory complex, thereby connecting translational regulation to actin dynamics and dendritic spine morphology, which are dysregulated in FXS model mice that lack FMRP. Treating FXS mice with 4EGI-1, which blocks interactions between eIF4E and eIF4G, a critical interaction partner for translational initiation, reversed defects in hippocampus-dependent memory and spine morphology. We also found that 4EGI-1 normalized the phenotypes of enhanced metabotropic glutamate receptor (mGluR)-mediated long-term depression (LTD), enhanced Rac1-p21-activated kinase (PAK)-cofilin signaling, altered actin dynamics, and dysregulated CYFIP1/eIF4E and CYFIP1/Rac1 interactions in FXS mice. Our findings are consistent with the idea that an imbalance in protein synthesis and actin dynamics contributes to pathophysiology in FXS mice, and suggest that targeting eIF4E may be a strategy for treating FXS.
Collapse
Affiliation(s)
- Emanuela Santini
- Center for Neural Science, New York University, New York, NY 10003, USA.,Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Thu N Huynh
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Francesco Longo
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - So Yeon Koo
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Edward Mojica
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Laura D'Andrea
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 00133 Rome, Italy
| | - Claudia Bagni
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 00133 Rome, Italy.,Center for Human Genetics and Leuven Research Institute for Neuroscience and Disease, KU Leuven, 3000 Leuven, Belgium.,VIB Center for the Biology of Disease, 3000 Leuven, Belgium.,Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland
| | - Eric Klann
- Center for Neural Science, New York University, New York, NY 10003, USA.
| |
Collapse
|
22
|
Levin N, Kritman M, Maroun M, Akirav I. Differential roles of the infralimbic and prelimbic areas of the prefrontal cortex in reconsolidation of a traumatic memory. Eur Neuropsychopharmacol 2017. [PMID: 28647452 DOI: 10.1016/j.euroneuro.2017.06.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Studies about reconsolidation of conditioned fear memories have shown that pharmacological manipulation at memory reactivation can attenuate or enhance the subsequent expression of the conditioned fear response. Here we examined the effects of a single injection of the mTOR inhibitor rapamycin (Rap) into the infralimbic (IL) and prelimbic (PL) areas [which compose the ventromedial prefrontal cortex (PFC)] on reconsolidation and extinction of a traumatic fear memory. We found opposite effects of Rap infused into the PL and IL on reconsolidation and extinction: intra-PL Rap and systemic Rap impaired reconsolidation and facilitated extinction whereas intra-IL Rap enhanced reconsolidation and impaired extinction. These effects persisted at least 10 days after reactivation. Shock exposure induced anxiety-like behavior and impaired working memory and intra-IL and -PL Rap normalized these effects. Finally, when measured after fear retrieval, shocked rats exhibited reduced and increased phosphorylated p70s6K levels in the IL and basolateral amygdala, respectively. No effect on phosphorylated p70s6K levels was observed in the PL. The study points to the differential roles of the IL and PL in memory reconsolidation and extinction. Moreover, inhibiting mTOR via rapamycin following reactivation of a fear memory may be a novel approach in attenuating enhanced fear memories.
Collapse
Affiliation(s)
- Natali Levin
- Department of Psychology, University of Haifa, Haifa 3498838, Israel
| | - Milly Kritman
- Sagol Department of Neurobiology, University of Haifa, Haifa 3498838, Israel
| | - Mouna Maroun
- Sagol Department of Neurobiology, University of Haifa, Haifa 3498838, Israel
| | - Irit Akirav
- Department of Psychology, University of Haifa, Haifa 3498838, Israel.
| |
Collapse
|
23
|
Roesler R. Molecular mechanisms controlling protein synthesis in memory reconsolidation. Neurobiol Learn Mem 2017; 142:30-40. [DOI: 10.1016/j.nlm.2017.04.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 04/27/2017] [Accepted: 04/28/2017] [Indexed: 10/19/2022]
|
24
|
McCamphill PK, Ferguson L, Sossin WS. A decrease in eukaryotic elongation factor 2 phosphorylation is required for local translation of sensorin and long-term facilitation in Aplysia. J Neurochem 2017; 142:246-259. [PMID: 28345161 DOI: 10.1111/jnc.14030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/15/2017] [Accepted: 03/17/2017] [Indexed: 12/20/2022]
Abstract
Mechanistic target of rapamycin complex 1 (mTORC1)-dependent protein synthesis is required for many forms of synaptic plasticity and memory, but the downstream pathways important for synaptic plasticity are poorly understood. Long-term facilitation (LTF) in Aplysia is a form of synaptic plasticity that is closely linked to behavioral memory and an attractive model system for examining the important downstream targets for mTORC1 in regulating synaptic plasticity. Although mTORC1-regulated protein synthesis has been strongly linked to translation initiation, translation elongation is also regulated by mTORC1 and LTF leads to an mTORC1-dependent decrease in eukaryotic elongation factor 2 (eEF2) phosphorylation. The purpose of this study is to test the hypothesis that the decrease in eEF2 phosphorylation is required for mTORC1-dependent translation and plasticity. We show that the LTF-induced decrease in eEF2 phosphorylation is blocked by expression of an eEF2 kinase (eEF2K) modified to be resistant to mTORC1 regulation. We found that expression of this modified kinase blocked LTF. LTF requires local protein synthesis of the neuropeptide sensorin and importantly, local sensorin synthesis can be measured using a dendra fluorescent protein containing the 5' and 3' untranslated regions (UTRs) of sensorin. Using this construct, we show that blocking eEF2 dephosphorylation also blocks the increase in local sensorin synthesis. These results identify decreases in eEF2 phosphorylation as a critical downstream effector of mTOR required for long-term plasticity and identify an important translational target regulated by decreases in eEF2 phosphorylation.
Collapse
Affiliation(s)
- Patrick K McCamphill
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Larissa Ferguson
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Wayne S Sossin
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
25
|
Huang WC, Chen Y, Page DT. Hyperconnectivity of prefrontal cortex to amygdala projections in a mouse model of macrocephaly/autism syndrome. Nat Commun 2016; 7:13421. [PMID: 27845329 PMCID: PMC5116076 DOI: 10.1038/ncomms13421] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 10/03/2016] [Indexed: 02/07/2023] Open
Abstract
Multiple autism risk genes converge on the regulation of mTOR signalling, which is a key effector of neuronal growth and connectivity. We show that mTOR signalling is dysregulated during early postnatal development in the cerebral cortex of germ-line heterozygous Pten mutant mice (Pten+/-), which model macrocephaly/autism syndrome. The basolateral amygdala (BLA) receives input from subcortical-projecting neurons in the medial prefrontal cortex (mPFC). Analysis of mPFC to BLA axonal projections reveals that Pten+/- mice exhibit increased axonal branching and connectivity, which is accompanied by increased activity in the BLA in response to social stimuli and social behavioural deficits. The latter two phenotypes can be suppressed by pharmacological inhibition of S6K1 during early postnatal life or by reducing the activity of mPFC-BLA circuitry in adulthood. These findings identify a mechanism of altered connectivity that has potential relevance to the pathophysiology of macrocephaly/autism syndrome and autism spectrum disorders featuring dysregulated mTOR signalling.
Collapse
Affiliation(s)
- Wen-Chin Huang
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA.,The Doctoral Program in Chemical and Biological Sciences, The Scripps Research Institute, USA
| | - Youjun Chen
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Damon T Page
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA.,The Doctoral Program in Chemical and Biological Sciences, The Scripps Research Institute, USA
| |
Collapse
|
26
|
Dopamine D2 receptors gate generalization of conditioned threat responses through mTORC1 signaling in the extended amygdala. Mol Psychiatry 2016; 21:1545-1553. [PMID: 26782052 PMCID: PMC5101541 DOI: 10.1038/mp.2015.210] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 10/28/2015] [Accepted: 11/19/2015] [Indexed: 12/17/2022]
Abstract
Overgeneralization of conditioned threat responses is a robust clinical marker of anxiety disorders. In overgeneralization, responses that are appropriate to threat-predicting cues are evoked by perceptually similar safety-predicting cues. Inappropriate learning of conditioned threat responses may thus form an etiological basis for anxiety disorders. The role of dopamine (DA) in memory encoding is well established. Indeed by signaling salience and valence, DA is thought to facilitate discriminative learning between stimuli representing safety or threat. However, the neuroanatomical and biochemical substrates through which DA modulates overgeneralization of threat responses remain poorly understood. Here we report that the modulation of DA D2 receptor (D2R) signaling bidirectionally regulates the consolidation of fear responses. While the blockade of D2R induces generalized threat responses, its stimulation facilitates discriminative learning between stimuli representing safety or threat. Moreover, we show that controlled threat generalization requires the coordinated activation of D2R in the bed nucleus of the stria terminalis and the central amygdala. Finally, we identify the mTORC1 cascade activation as an important molecular event by which D2R mediates its effects. These data reveal that D2R signaling in the extended amygdala constitutes an important checkpoint through which DA participates in the control of threat processing and the emergence of overgeneralized threat responses.
Collapse
|
27
|
Targeting Translation Control with p70 S6 Kinase 1 Inhibitors to Reverse Phenotypes in Fragile X Syndrome Mice. Neuropsychopharmacology 2016; 41:1991-2000. [PMID: 26708105 PMCID: PMC4908636 DOI: 10.1038/npp.2015.369] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 12/11/2015] [Accepted: 12/22/2015] [Indexed: 11/08/2022]
Abstract
Aberrant neuronal translation is implicated in the etiology of numerous brain disorders. Although mTORC1-p70 ribosomal S6 kinase 1 (S6K1) signaling is critical for translational control, pharmacological manipulation in vivo has targeted exclusively mTORC1 due to the paucity of specific inhibitors to S6K1. However, small molecule inhibitors of S6K1 could potentially ameliorate pathological phenotypes of diseases, which are based on aberrant translation and protein expression. One such condition is fragile X syndrome (FXS), which is considered to be caused by exaggerated neuronal translation and is the most frequent heritable cause of autism spectrum disorder (ASD). To date, potential therapeutic interventions in FXS have focused largely on targets upstream of translational control to normalize FXS-related phenotypes. Here we test the ability of two S6K1 inhibitors, PF-4708671 and FS-115, to normalize translational homeostasis and other phenotypes exhibited by FXS model mice. We found that although the pharmacokinetic profiles of the two S6K1 inhibitors differed, they overlapped in reversing multiple disease-associated phenotypes in FXS model mice including exaggerated protein synthesis, inappropriate social behavior, behavioral inflexibility, altered dendritic spine morphology, and macroorchidism. In contrast, the two inhibitors differed in their ability to rescue stereotypic marble-burying behavior and weight gain. These findings provide an initial pharmacological characterization of the impact of S6K1 inhibitors in vivo for FXS, and have therapeutic implications for other neuropsychiatric conditions involving aberrant mTORC1-S6K1 signaling.
Collapse
|
28
|
Persistent Associative Plasticity at an Identified Synapse Underlying Classical Conditioning Becomes Labile with Short-Term Homosynaptic Activation. J Neurosci 2016; 35:16159-70. [PMID: 26658867 DOI: 10.1523/jneurosci.2034-15.2015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Synapses express different forms of plasticity that contribute to different forms of memory, and both memory and plasticity can become labile after reactivation. We previously reported that a persistent form of nonassociative long-term facilitation (PNA-LTF) of the sensorimotor synapses in Aplysia californica, a cellular analog of long-term sensitization, became labile with short-term heterosynaptic reactivation and reversed when the reactivation was followed by incubation with the protein synthesis inhibitor rapamycin. Here we examined the reciprocal impact of different forms of short-term plasticity (reactivations) on a persistent form of associative long-term facilitation (PA-LTF), a cellular analog of classical conditioning, which was expressed at Aplysia sensorimotor synapses when a tetanic stimulation of the sensory neurons was paired with a brief application of serotonin on 2 consecutive days. The expression of short-term homosynaptic plasticity [post-tetanic potentiation or homosynaptic depression (HSD)], or short-term heterosynaptic plasticity [serotonin-induced facilitation or neuropeptide Phe-Met-Arg-Phe-NH2 (FMRFa)-induced depression], at synapses expressing PA-LTF did not affect the maintenance of PA-LTF. The kinetics of HSD was attenuated at synapses expressing PA-LTF, which required activation of protein kinase C (PKC). Both PA-LTF and the attenuated kinetics of HSD were reversed by either a transient blockade of PKC activity or a homosynaptic, but not heterosynaptic, reactivation when paired with rapamycin. These results indicate that two different forms of persistent synaptic plasticity, PA-LTF and PNA-LTF, expressed at the same synapse become labile when reactivated by different stimuli. SIGNIFICANCE STATEMENT Activity-dependent changes in neural circuits mediate long-term memories. Some forms of long-term memories become labile and can be reversed with specific types of reactivations, but the mechanism is complex. At the cellular level, reactivations that induce a reversal of memory must evoke changes in neural circuits underlying the memory. What types of reactivations induce a labile state at neural connections that lead to reversal of different types of memory? We find that a critical neural connection in Aplysia, which is modified with different stimuli that mediate different types of memory, becomes labile with different types of reactivations. These results provide insights for developing strategies in alleviating maladaptive memories accompanying anxiety disorders.
Collapse
|
29
|
Hill JL, Martinowich K. Activity-dependent signaling: influence on plasticity in circuits controlling fear-related behavior. Curr Opin Neurobiol 2016; 36:59-65. [PMID: 26485574 PMCID: PMC4738053 DOI: 10.1016/j.conb.2015.10.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 09/30/2015] [Accepted: 10/01/2015] [Indexed: 10/22/2022]
Abstract
Fear regulation is impaired in anxiety and trauma-related disorders. Patients experience heightened fear expression and reduced ability to extinguish fear memories. Because fear regulation is abnormal in these disorders and extinction recapitulates current treatment strategies, understanding the underlying mechanisms is vital for developing new treatments. This is critical because although extinction-based exposure therapy is a mainstay of treatment, relapse is common. We examine recent findings describing changes in network activity and functional connectivity within limbic circuits during fear regulation, and explore how activity-dependent signaling contributes to the neural activity patterns that control fear and anxiety. We review the role of the prototypical activity-dependent molecule, brain-derived neurotrophic factor (BDNF), whose signaling has been critically linked to regulation of fear behavior.
Collapse
Affiliation(s)
- Julia L Hill
- Lieber Institute for Brain Development, Baltimore, MD 21205, United States
| | - Keri Martinowich
- Lieber Institute for Brain Development, Baltimore, MD 21205, United States; Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, United States.
| |
Collapse
|
30
|
Huynh TN, Shah M, Koo SY, Faraud KS, Santini E, Klann E. eIF4E/Fmr1 double mutant mice display cognitive impairment in addition to ASD-like behaviors. Neurobiol Dis 2015; 83:67-74. [PMID: 26306459 DOI: 10.1016/j.nbd.2015.08.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 07/23/2015] [Accepted: 08/17/2015] [Indexed: 11/25/2022] Open
Abstract
Autism spectrum disorder (ASD) is a group of heritable disorders with complex and unclear etiology. Classic ASD symptoms include social interaction and communication deficits as well as restricted, repetitive behaviors. In addition, ASD is often comorbid with intellectual disability. Fragile X syndrome (FXS) is the leading genetic cause of ASD, and is the most commonly inherited form of intellectual disability. Several mouse models of ASD and FXS exist, however the intellectual disability observed in ASD patients is not well modeled in mice. Using the Fmr1 knockout mouse and the eIF4E transgenic mouse, two previously characterized mouse models of fragile X syndrome and ASD, respectively, we generated the eIF4E/Fmr1 double mutant mouse. Our study shows that the eIF4E/Fmr1 double mutant mice display classic ASD behaviors, as well as cognitive dysfunction. Importantly, the learning impairments displayed by the double mutant mice spanned multiple cognitive tasks. Moreover, the eIF4E/Fmr1 double mutant mice display increased levels of basal protein synthesis. The results of our study suggest that the eIF4E/Fmr1 double mutant mouse may be a reliable model to study cognitive dysfunction in the context of ASD.
Collapse
Affiliation(s)
- Thu N Huynh
- Center for Neural Science, New York University, New York, NY 10003, United States
| | - Manan Shah
- Center for Neural Science, New York University, New York, NY 10003, United States
| | - So Yeon Koo
- Center for Neural Science, New York University, New York, NY 10003, United States
| | - Kirsten S Faraud
- Center for Neural Science, New York University, New York, NY 10003, United States
| | - Emanuela Santini
- Center for Neural Science, New York University, New York, NY 10003, United States
| | - Eric Klann
- Center for Neural Science, New York University, New York, NY 10003, United States
| |
Collapse
|
31
|
Perl A. mTOR activation is a biomarker and a central pathway to autoimmune disorders, cancer, obesity, and aging. Ann N Y Acad Sci 2015; 1346:33-44. [PMID: 25907074 DOI: 10.1111/nyas.12756] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The mechanistic target of rapamycin (mTOR) is a ubiquitous serine/threonine kinase, which plays pivotal roles in integrating growth signals on a cellular level. To support proliferation and survival under stress, two interacting complexes that harbor mTOR, mTORC1 and mTORC2, promote the transcription of genes involved in carbohydrate metabolism and lipogenesis, enhance protein translation, and inhibit autophagy. Although rapamycin was originally developed as an inhibitor of T cell proliferation for preventing organ transplant rejection, its molecular target, mTOR, has been subsequently identified as a central regulator of metabolic cues that drive lineage specification in the immune system. Owing to oxidative stress, the activation of mTORC1 has emerged as a central pathway for the pathogenesis of systemic lupus erythematosus and other autoimmune diseases. Paradoxically, mTORC1 has also been identified as a mediator of the Warburg effect that allows cell survival under hypoxia. Rapamycin and new classes of mTOR inhibitors are being developed to block not only transplant rejection and autoimmunity but also to treat obesity and various forms of cancer. Through preventing these diseases, personalized mTOR blockade holds promise to extend life span.
Collapse
Affiliation(s)
- Andras Perl
- Division of Rheumatology, Department of Medicine State University of New York, Upstate Medical University, College of Medicine, Syracuse, New York.,Division of Rheumatology, Department of Microbiology and Immunology, State University of New York, Upstate Medical University, College of Medicine, Syracuse, New York
| |
Collapse
|
32
|
Mérour JY, Buron F, Plé K, Bonnet P, Routier S. The azaindole framework in the design of kinase inhibitors. Molecules 2014; 19:19935-79. [PMID: 25460315 PMCID: PMC6271083 DOI: 10.3390/molecules191219935] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 11/10/2014] [Accepted: 11/18/2014] [Indexed: 01/05/2023] Open
Abstract
This review article illustrates the growing use of azaindole derivatives as kinase inhibitors and their contribution to drug discovery and innovation. The different protein kinases which have served as targets and the known molecules which have emerged from medicinal chemistry and Fragment-Based Drug Discovery (FBDD) programs are presented. The various synthetic routes used to access these compounds and the chemical pathways leading to their synthesis are also discussed. An analysis of their mode of binding based on X-ray crystallography data gives structural insights for the design of more potent and selective inhibitors.
Collapse
Affiliation(s)
- Jean-Yves Mérour
- Institut de Chimie Organique et Analytique (ICOA), Université d'Orléans, UMR CNRS 7311, Orléans F-45067, France.
| | - Frédéric Buron
- Institut de Chimie Organique et Analytique (ICOA), Université d'Orléans, UMR CNRS 7311, Orléans F-45067, France.
| | - Karen Plé
- Institut de Chimie Organique et Analytique (ICOA), Université d'Orléans, UMR CNRS 7311, Orléans F-45067, France.
| | - Pascal Bonnet
- Institut de Chimie Organique et Analytique (ICOA), Université d'Orléans, UMR CNRS 7311, Orléans F-45067, France.
| | - Sylvain Routier
- Institut de Chimie Organique et Analytique (ICOA), Université d'Orléans, UMR CNRS 7311, Orléans F-45067, France.
| |
Collapse
|