1
|
Mossa A, Dierdorff L, Lukin J, Garcia-Forn M, Wang W, Mamashli F, Park Y, Fiorenzani C, Akpinar Z, Kamps J, Tatzelt J, Wu Z, De Rubeis S. Sex-specific perturbations of neuronal development caused by mutations in the autism risk gene DDX3X. Nat Commun 2025; 16:4512. [PMID: 40374608 PMCID: PMC12081640 DOI: 10.1038/s41467-025-59680-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 04/29/2025] [Indexed: 05/17/2025] Open
Abstract
DDX3X is an X-linked RNA helicase that escapes X chromosome inactivation and is expressed at higher levels in female brains. Mutations in DDX3X are associated with intellectual disability (ID) and autism spectrum disorder (ASD) and are predominantly identified in females (DDX3X syndrome). Using cellular and mouse models, we show that Ddx3x mediates sexual dimorphisms in brain development at a molecular, cellular, and behavioral level. During cortical neuronal development, Ddx3x sustains a female-biased signature of enhanced ribosomal biogenesis and mRNA metabolism. Compared to male neurons, female neurons display larger nucleoli, higher expression of a set of ribosomal proteins, and a higher cytoplasm-to-nucleus ratio of ribosomal RNA. All these sex dimorphisms are obliterated by Ddx3x loss. Ddx3x regulates dendritic arborization complexity in a sex- and dose-dependent manner in both female and male neurons. Ddx3x modulates the development of dendritic spines but only in female neurons. Further, ablating Ddx3x conditionally in forebrain neurons is sufficient to yield sex-specific changes in developmental outcomes and motor function. Together, these findings pose Ddx3x as a mediator of sexual differentiation during neurodevelopment and open new avenues to understand sex differences in health and disease.
Collapse
Affiliation(s)
- Adele Mossa
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Alper Center for Neural Development and Regeneration Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Lauren Dierdorff
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Alper Center for Neural Development and Regeneration Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jeronimo Lukin
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Alper Center for Neural Development and Regeneration Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Marta Garcia-Forn
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Alper Center for Neural Development and Regeneration Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Wei Wang
- Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY10021, USA
| | - Fatemeh Mamashli
- Department Biochemistry of Neurodegenerative Diseases, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Yeaji Park
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Alper Center for Neural Development and Regeneration Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Chiara Fiorenzani
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Alper Center for Neural Development and Regeneration Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Zeynep Akpinar
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Alper Center for Neural Development and Regeneration Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Biology, New York University, College of Arts and Science, New York, NY, 10003, USA
| | - Janine Kamps
- Department Biochemistry of Neurodegenerative Diseases, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Jörg Tatzelt
- Department Biochemistry of Neurodegenerative Diseases, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
- Cluster of Excellence RESOLV, Bochum, Germany
| | - Zhuhao Wu
- Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY10021, USA
| | - Silvia De Rubeis
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Alper Center for Neural Development and Regeneration Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
2
|
Trent S, Abdullah MH, Parwana K, Valdivieso MA, Hassan Z, Müller CP. Fear conditioning: Insights into learning, memory and extinction and its relevance to clinical disorders. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111310. [PMID: 40056965 DOI: 10.1016/j.pnpbp.2025.111310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/26/2025] [Accepted: 03/02/2025] [Indexed: 03/15/2025]
Abstract
Fear, whether innate or learned, is an essential emotion required for survival. The learning, and subsequent memory, of fearful events enhances our ability to recognise and respond to threats, aiding adaptation to new, ever-changing environments. Considerable research has leveraged associative learning protocols such as contextual or auditory forms of fear conditioning in rodents, to understand fear learning, memory consolidation and extinction phases of memory. Such assays have led to detailed characterisation of the underlying neurocircuitry and neurobiology supporting fear learning processes. Given fear processing is conserved across rodents and humans, fear conditioning experiments provide translational insights into fundamental memory processes and fear-related pathologies. This review examines associative learning protocols used to measure fear learning, memory and extinction, before providing an overview on the underlying complex neurocircuitry including the amygdala, hippocampus and medial prefrontal cortex. This is followed by an in-depth commentary on the neurobiology, particularly synaptic plasticity mechanisms, which regulate fear learning, memory and extinction. Next, we consider how fear conditioning assays in rodents can inform our understanding of disrupted fear memory in human disorders such as post-traumatic stress disorder (PTSD), anxiety and psychiatric disorders including schizophrenia. Lastly, we critically evaluate fear conditioning protocols, highlighting some of the experimental and theoretical limitations and the considerations required when conducting such assays, alongside recent methodological advancements in the field. Overall, rodent-based fear conditioning assays remain central to making progress in uncovering fundamental memory phenomena and understanding the aetiological mechanisms that underpin fear associated disorders, alongside the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Simon Trent
- School of Life Sciences, Faculty of Natural Sciences, Huxley Building, Keele University, Keele ST5 5BG, UK.
| | | | - Krishma Parwana
- School of Life Sciences, Faculty of Natural Sciences, Huxley Building, Keele University, Keele ST5 5BG, UK
| | - Maria Alcocer Valdivieso
- School of Life Sciences, Faculty of Natural Sciences, Huxley Building, Keele University, Keele ST5 5BG, UK
| | - Zurina Hassan
- Centre for Drug Research, Universiti Malaysia (USM), 11800 Penang, Malaysia
| | - Christian P Müller
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, 91054 Erlangen, Germany; Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
3
|
Henis M, Rücker T, Scharrenberg R, Richter M, Baltussen L, Hong S, Meka DP, Schwanke B, Neelagandan N, Daaboul D, Murtaza N, Krisp C, Harder S, Schlüter H, Kneussel M, Hermans-Borgmeyer I, de Wit J, Singh KK, Duncan KE, de Anda FC. The autism susceptibility kinase, TAOK2, phosphorylates eEF2 and modulates translation. SCIENCE ADVANCES 2024; 10:eadf7001. [PMID: 38608030 PMCID: PMC11014455 DOI: 10.1126/sciadv.adf7001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 03/12/2024] [Indexed: 04/14/2024]
Abstract
Genes implicated in translation control have been associated with autism spectrum disorders (ASDs). However, some important genetic causes of autism, including the 16p11.2 microdeletion, bear no obvious connection to translation. Here, we use proteomics, genetics, and translation assays in cultured cells and mouse brain to reveal altered translation mediated by loss of the kinase TAOK2 in 16p11.2 deletion models. We show that TAOK2 associates with the translational machinery and functions as a translational brake by phosphorylating eukaryotic elongation factor 2 (eEF2). Previously, all signal-mediated regulation of translation elongation via eEF2 phosphorylation was believed to be mediated by a single kinase, eEF2K. However, we show that TAOK2 can directly phosphorylate eEF2 on the same regulatory site, but functions independently of eEF2K signaling. Collectively, our results reveal an eEF2K-independent signaling pathway for control of translation elongation and suggest altered translation as a molecular component in the etiology of some forms of ASD.
Collapse
Affiliation(s)
- Melad Henis
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, New Valley University, 72511 El-Kharga, Egypt
| | - Tabitha Rücker
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Robin Scharrenberg
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Melanie Richter
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Lucas Baltussen
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium
- KU Leuven Department of Neurosciences, Leuven Brain Institute, Herestraat 49, 3000 Leuven, Belgium
| | - Shuai Hong
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Durga Praveen Meka
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Birgit Schwanke
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Nagammal Neelagandan
- Neuronal Translational Control Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Falkenried 94, 20251 Hamburg, Germany
- Institute of Bioengineering (IBI), École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Danie Daaboul
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium
- KU Leuven Department of Neurosciences, Leuven Brain Institute, Herestraat 49, 3000 Leuven, Belgium
| | - Nadeem Murtaza
- Krembil Research Institute, Donald K. Johnson Eye Institute, University Health Network, 60 Leonard Ave, Toronto, Ontario M5T 0S8, Canada
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, Ontario L8S 4A9, Canada
| | - Christoph Krisp
- Institute for Clinical Chemistry and Laboratory Medicine, Mass Spectrometric Proteomics Group, Campus Forschung, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany
| | - Sönke Harder
- Institute for Clinical Chemistry and Laboratory Medicine, Mass Spectrometric Proteomics Group, Campus Forschung, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany
| | - Hartmut Schlüter
- Institute for Clinical Chemistry and Laboratory Medicine, Mass Spectrometric Proteomics Group, Campus Forschung, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany
| | - Matthias Kneussel
- Institute of Neurogenetics, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf (UKE), 20251 Hamburg, Germany
| | - Irm Hermans-Borgmeyer
- Transgenic Service Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Falkenried 94, 20251 Hamburg, Germany
| | - Joris de Wit
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium
- KU Leuven Department of Neurosciences, Leuven Brain Institute, Herestraat 49, 3000 Leuven, Belgium
| | - Karun K. Singh
- Krembil Research Institute, Donald K. Johnson Eye Institute, University Health Network, 60 Leonard Ave, Toronto, Ontario M5T 0S8, Canada
- Faculty of Medicine, University of Toronto, Medical Sciences Building, 1 King's College Cir, Toronto, Ontario M5S 1 A8, Canada
| | - Kent E. Duncan
- Neuronal Translational Control Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Falkenried 94, 20251 Hamburg, Germany
- Evotec SE, Manfred Eigen Campus, Essener Bogen 7, 22419 Hamburg, Germany
| | - Froylan Calderón de Anda
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
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
|
Longo F, Aryal S, Anastasiades PG, Maltese M, Baimel C, Albanese F, Tabor J, Zhu JD, Oliveira MM, Gastaldo D, Bagni C, Santini E, Tritsch NX, Carter AG, Klann E. Cell-type-specific disruption of cortico-striatal circuitry drives repetitive patterns of behavior in fragile X syndrome model mice. Cell Rep 2023; 42:112901. [PMID: 37505982 PMCID: PMC10552611 DOI: 10.1016/j.celrep.2023.112901] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 05/18/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Individuals with fragile X syndrome (FXS) are frequently diagnosed with autism spectrum disorder (ASD), including increased risk for restricted and repetitive behaviors (RRBs). Consistent with observations in humans, FXS model mice display distinct RRBs and hyperactivity that are consistent with dysfunctional cortico-striatal circuits, an area relatively unexplored in FXS. Using a multidisciplinary approach, we dissect the contribution of two populations of striatal medium spiny neurons (SPNs) in the expression of RRBs in FXS model mice. Here, we report that dysregulated protein synthesis at cortico-striatal synapses is a molecular culprit of the synaptic and ASD-associated motor phenotypes displayed by FXS model mice. Cell-type-specific translational profiling of the FXS mouse striatum reveals differentially translated mRNAs, providing critical information concerning potential therapeutic targets. Our findings uncover a cell-type-specific impact of the loss of fragile X messenger ribonucleoprotein (FMRP) on translation and the sequence of neuronal events in the striatum that drive RRBs in FXS.
Collapse
Affiliation(s)
- Francesco Longo
- Center for Neural Science, New York University, New York, NY 10003, USA; Institute for Neuroscience and Physiology, University of Gothenburg, 40530 Gothenburg, Sweden; Sackler Institute of Graduate Biomedical Sciences, NYU School of Medicine, New York, NY 10016, USA
| | - Sameer Aryal
- Center for Neural Science, New York University, New York, NY 10003, USA; NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, USA
| | | | - Marta Maltese
- Fresco Institute for Parkinson's and Movement Disorders, New York University Langone Health, New York, NY 10016, USA; Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland
| | - Corey Baimel
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Federica Albanese
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Joanna Tabor
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Jeffrey D Zhu
- Center for Neural Science, New York University, New York, NY 10003, USA
| | | | - Denise Gastaldo
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 1005 Rome, Italy
| | - Claudia Bagni
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata," 1005 Rome, Italy
| | - Emanuela Santini
- Center for Neural Science, New York University, New York, NY 10003, USA; Department of Neuroscience, Biomedicum, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Nicolas X Tritsch
- NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, USA; Fresco Institute for Parkinson's and Movement Disorders, New York University Langone Health, New York, NY 10016, USA
| | - Adam G Carter
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Eric Klann
- Center for Neural Science, New York University, New York, NY 10003, USA; NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
7
|
Lopes PC, Faber-Hammond JJ, Siemonsma C, Patel S, Renn SCP. The social environment alters neural responses to a lipopolysaccharide challenge. Brain Behav Immun 2023; 110:162-174. [PMID: 36878331 DOI: 10.1016/j.bbi.2023.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Sick animals display drastic changes in their behavioral patterns, including decreased activity, decreased food and water intake, and decreased interest in social interactions. These behaviors, collectively called "sickness behaviors", can be socially modulated. For example, when provided with mating opportunities, males of several species show reduced sickness behaviors. While the behavior is known to change, how the social environment affects neural molecular responses to sickness is not known. Here, we used a species, the zebra finch, Taeniopygia guttata, where males have been shown to decrease sickness behaviors when presented with novel females. Using this paradigm, we obtained samples from three brain regions (the hypothalamus, the bed nucleus of the stria terminalis, and the nucleus taeniae) from lipopolysaccharide (LPS) or control treated males housed under four different social environments. Manipulation of the social environment rapidly changed the strength and co-expression patterns of the neural molecular responses to the immune challenge in all brain regions tested, therefore suggesting that the social environment plays a significant role in determining the neural responses to an infection. In particular, brains of males paired with a novel female showed muted immune responses to LPS, as well as altered synaptic signaling. Neural metabolic activity in response to the LPS challenge was also affected by the social environment. Our results provide new insights into the effects of the social environment on brain responses to an infection, thereby improving our understanding of how the social environment can affect health.
Collapse
Affiliation(s)
- Patricia C Lopes
- Schmid College of Science and Technology, Chapman University, Orange, CA, USA.
| | | | - Chandler Siemonsma
- Schmid College of Science and Technology, Chapman University, Orange, CA, USA
| | - Sachin Patel
- Schmid College of Science and Technology, Chapman University, Orange, CA, USA
| | - Suzy C P Renn
- Department of Biology, Reed College, Portland, OR, USA
| |
Collapse
|
8
|
Wiebe S, Huang Z, Ladak RJ, Skalecka A, Cagnetta R, Lacaille JC, Aguilar-Valles A, Sonenberg N. Cell-type-specific translational control of spatial working memory by the cap-binding protein 4EHP. Mol Brain 2023; 16:9. [PMID: 36650535 PMCID: PMC9847188 DOI: 10.1186/s13041-023-00995-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 01/01/2023] [Indexed: 01/19/2023] Open
Abstract
The consolidation of learned information into long-lasting memories requires the strengthening of synaptic connections through de novo protein synthesis. Translation initiation factors play a cardinal role in gating the production of new proteins thereby regulating memory formation. Both positive and negative regulators of translation play a critical role in learning and memory consolidation. The eukaryotic initiation factor 4E (eIF4E) homologous protein (4EHP, encoded by the gene Eif4e2) is a pivotal negative regulator of translation but its role in learning and memory is unknown. To address this gap in knowledge, we generated excitatory (glutamatergic: CaMKIIα-positive) and inhibitory (GABAergic: GAD65-positive) conditional knockout mice for 4EHP, which were analyzed in various behavioral memory tasks. Knockout of 4EHP in Camk2a-expressing neurons (4EHP-cKOexc) did not impact long-term memory in either contextual fear conditioning or Morris water maze tasks. Similarly, long-term contextual fear memory was not altered in Gad2-directed 4EHP knockout mice (4EHP-cKOinh). However, when subjected to a short-term T-maze working memory task, both mouse models exhibited impaired cognition. We therefore tested the hypothesis that de novo protein synthesis plays a direct role in working memory. We discovered that phosphorylation of ribosomal protein S6, a measure of mTORC1 activity, is dramatically reduced in the CA1 hippocampus of 4EHP-cKOexc mice. Consistently, genetic reduction of mTORC1 activity in either excitatory or inhibitory neurons was sufficient to impair working memory. Taken together, these findings indicate that translational control by 4EHP and mTORC1 in both excitatory and inhibitory neurons are necessary for working memory.
Collapse
Affiliation(s)
- Shane Wiebe
- grid.14709.3b0000 0004 1936 8649Department of Biochemistry, McGill University, McIntyre Medical Building, 3655 Promenade Sir William Osler, Montreal, QC H3G 1Y6 Canada ,Goodman Cancer Institute, 1160 Pine Avenue West, Room 614, Montreal, QC H3A 1A3 Canada
| | - Ziying Huang
- grid.14709.3b0000 0004 1936 8649Department of Biochemistry, McGill University, McIntyre Medical Building, 3655 Promenade Sir William Osler, Montreal, QC H3G 1Y6 Canada ,Goodman Cancer Institute, 1160 Pine Avenue West, Room 614, Montreal, QC H3A 1A3 Canada
| | - Reese Jalal Ladak
- grid.14709.3b0000 0004 1936 8649Department of Biochemistry, McGill University, McIntyre Medical Building, 3655 Promenade Sir William Osler, Montreal, QC H3G 1Y6 Canada ,Goodman Cancer Institute, 1160 Pine Avenue West, Room 614, Montreal, QC H3A 1A3 Canada
| | - Agnieszka Skalecka
- grid.14709.3b0000 0004 1936 8649Department of Biochemistry, McGill University, McIntyre Medical Building, 3655 Promenade Sir William Osler, Montreal, QC H3G 1Y6 Canada ,Goodman Cancer Institute, 1160 Pine Avenue West, Room 614, Montreal, QC H3A 1A3 Canada
| | - Roberta Cagnetta
- grid.14709.3b0000 0004 1936 8649Department of Biochemistry, McGill University, McIntyre Medical Building, 3655 Promenade Sir William Osler, Montreal, QC H3G 1Y6 Canada ,Goodman Cancer Institute, 1160 Pine Avenue West, Room 614, Montreal, QC H3A 1A3 Canada
| | - Jean-Claude Lacaille
- grid.14848.310000 0001 2292 3357Department of Neuroscience and CIRCA, University of Montreal, Succ. Downtown, P. O. Box 6128, Montreal, QC H3C 3J7 Canada
| | - Argel Aguilar-Valles
- grid.34428.390000 0004 1936 893XDepartment of Neuroscience, Carleton University, Health Sciences Building, 1125 Colonel By Drive, Ottawa, ON K1S 5B6 Canada
| | - Nahum Sonenberg
- grid.14709.3b0000 0004 1936 8649Department of Biochemistry, McGill University, McIntyre Medical Building, 3655 Promenade Sir William Osler, Montreal, QC H3G 1Y6 Canada ,Goodman Cancer Institute, 1160 Pine Avenue West, Room 614, Montreal, QC H3A 1A3 Canada
| |
Collapse
|
9
|
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
|
10
|
Ostroff LE, Cain CK. Persistent up-regulation of polyribosomes at synapses during long-term memory, reconsolidation, and extinction of associative memory. LEARNING & MEMORY (COLD SPRING HARBOR, N.Y.) 2022; 29:192-202. [PMID: 35882501 PMCID: PMC9374273 DOI: 10.1101/lm.053577.122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/28/2022] [Indexed: 11/25/2022]
Abstract
Local protein synthesis at synapses can provide a rapid supply of proteins to support synaptic changes during consolidation of new memories, but its role in the maintenance or updating of established memories is unknown. Consolidation requires new protein synthesis in the period immediately following learning, whereas established memories are resistant to protein synthesis inhibitors. We have previously reported that polyribosomes are up-regulated in the lateral amygdala (LA) during consolidation of aversive-cued Pavlovian conditioning. In this study, we used serial section electron microscopy reconstructions to determine whether the distribution of dendritic polyribosomes returns to baseline during the long-term memory phase. Relative to control groups, long-term memory was associated with up-regulation of polyribosomes throughout dendrites, including in dendritic spines of all sizes. Retrieval of a consolidated memory by presentation of a small number of cues induces a new, transient requirement for protein synthesis to maintain the memory, while presentation of a large number of cues results in extinction learning, forming a new memory. One hour after retrieval or extinction training, the distribution of dendritic polyribosomes was similar except in the smallest spines, which had more polyribosomes in the extinction group. Our results demonstrate that the effects of learning on dendritic polyribosomes are not restricted to the transient translation-dependent phase of memory formation. Cued Pavlovian conditioning induces persistent synapse strengthening in the LA that is not reversed by retrieval or extinction, and dendritic polyribosomes may therefore correlate generally with synapse strength as opposed to recent activity or transient translational processes.
Collapse
Affiliation(s)
- Linnaea E Ostroff
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut 06269, USA.,Connecticut Institute for the Brain and Cognitive Science, University of Connecticut, Storrs, Connecticut 06269, USA.,Institute of Materials Science, University of Connecticut, Storrs, Connecticut 06269, USA
| | - Christopher K Cain
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, New York 10962, USA.,Child and Adolescent Psychiatry, New York University Langone Health, New York, New York 10016, USA
| |
Collapse
|
11
|
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
|
12
|
van 't Spijker HM, Stackpole EE, Almeida S, Katsara O, Liu B, Shen K, Schneider RJ, Gao FB, Richter JD. Ribosome profiling reveals novel regulation of C9ORF72 GGGGCC repeat-containing RNA translation. RNA (NEW YORK, N.Y.) 2022; 28:123-138. [PMID: 34848561 PMCID: PMC8906550 DOI: 10.1261/rna.078963.121] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/31/2021] [Indexed: 06/13/2023]
Abstract
GGGGCC (G4C2) repeat expansion in the first intron of C9ORF72 causes amyotrophic lateral sclerosis and frontotemporal dementia. Repeat-containing RNA is translated into dipeptide repeat (DPR) proteins, some of which are neurotoxic. Using dynamic ribosome profiling, we identified three translation initiation sites in the intron upstream of (G4C2) repeats; these sites are detected irrespective of the presence or absence of the repeats. During translocation, ribosomes appear to be stalled on the repeats. An AUG in the preceding C9ORF72 exon initiates a uORF that inhibits downstream translation. Polysome isolation indicates that unspliced (G4C2) repeat-containing RNA is a substrate for DPR protein synthesis. (G4C2) repeat-containing RNA translation is 5' cap-independent but inhibited by the initiation factor DAP5, suggesting an interplay with uORF function. These results define novel translational mechanisms of expanded (G4C2) repeat-containing RNA in disease.
Collapse
Affiliation(s)
- Heleen M van 't Spijker
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA
| | - Emily E Stackpole
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA
| | - Sandra Almeida
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA
| | - Olga Katsara
- Department of Microbiology, New York University School of Medicine, New York, New York 10016, USA
| | - Botao Liu
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA
| | - Kuang Shen
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA
| | - Robert J Schneider
- Department of Microbiology, New York University School of Medicine, New York, New York 10016, USA
| | - Fen-Biao Gao
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA
| | - Joel D Richter
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA
| |
Collapse
|
13
|
Novel role of CAP1 in regulation RNA polymerase II-mediated transcription elongation depends on its actin-depolymerization activity in nucleoplasm. Oncogene 2021; 40:3492-3509. [PMID: 33911205 DOI: 10.1038/s41388-021-01789-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 03/21/2021] [Accepted: 04/12/2021] [Indexed: 02/02/2023]
Abstract
Lung cancer is one of the most intractable diseases with high incidence and mortality worldwide. Adenylate cyclase-associated protein 1 (CAP1), a well-known actin depolymerization factor, is recently reported to be an oncogene accelerating cancer cell proliferation. However, the physiological significance of CAP1 in lung cancer is incompletely understood and the novel functions of CAP1 in transcriptional regulation remain unknown. Here we found that CAP1 was highly expressed in lung cancer tissues and cells, which was also negatively associated with prognosis in lung cancer patients. Moreover, CAP1 promoted A549 cells proliferation by promoting protein synthesis to accelerate cell cycle progression. Mechanistically, we revealed that CAP1 facilitated cyclin-dependent kinase 9 (CDK9)-mediated RNA polymerases (Pol) II-Ser2 phosphorylation and subsequent transcription elongation, and CAP1 performed its function in this progress depending on its actin-depolymerization activity in nucleoplasm. Furthermore, our in vivo findings confirmed that CAP1-promoted A549 xenograft tumor growth was associated with CDK9-mediated Pol II-Ser2 phosphorylation. Our study elucidates a novel role of CAP1 in modulating transcription by promoting polymerase II phosphorylation and suggests that CAP1 is a newly identified biomarker for lung cancer treatment and prognosis prediction.
Collapse
|
14
|
Gindina S, Botsford B, Cowansage K, LeDoux J, Klann E, Hoeffer C, Ostroff L. Upregulation of eIF4E, but not other translation initiation factors, in dendritic spines during memory formation. J Comp Neurol 2021; 529:3112-3126. [PMID: 33864263 DOI: 10.1002/cne.25158] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/27/2021] [Accepted: 04/11/2021] [Indexed: 11/09/2022]
Abstract
Local translation can provide a rapid, spatially targeted supply of new proteins in distal dendrites to support synaptic changes that underlie learning. Learning and memory are especially sensitive to manipulations of translational control mechanisms, particularly those that target the initiation step, and translation initiation at synapses could be a means of maintaining synapse specificity during plasticity. Initiation predominantly occurs via recruitment of ribosomes to the 5' mRNA cap by complexes of eukaryotic initiation factors (eIFs), and the interaction between eIF4E and eIF4G1 is a particularly important target of translational control pathways. Pharmacological inhibition of eIF4E-eIF4G1 binding impairs formation of memory for aversive Pavlovian conditioning as well as the accompanying increase in polyribosomes in the heads of dendritic spines in the lateral amygdala (LA). This is consistent with a role for initiation at synapses in memory formation, but whether eIFs are even present near synapses is unknown. To determine whether dendritic spines contain eIFs and whether eIF distribution is affected by learning, we combined immunolabeling with serial section transmission electron microscopy (ssTEM) volume reconstructions of LA dendrites after Pavlovian conditioning. Labeling for eIF4E, eIF4G1, and eIF2α-another key target of regulation-occurred in roughly half of dendritic spines, but learning effects were only found for eIF4E, which was upregulated in the heads of dendritic spines. Our results support the possibility of regulated translation initiation as a means of synapse-specific protein targeting during learning and are consistent with the model of eIF4E availability as a central point of control.
Collapse
Affiliation(s)
- Sofya Gindina
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York, USA
| | - Benjamin Botsford
- Center for Neural Science, New York University, New York, New York, USA
| | - Kiriana Cowansage
- Center for Neural Science, New York University, New York, New York, USA
| | - Joseph LeDoux
- Center for Neural Science, New York University, New York, New York, USA.,Nathan Kline Institute for Psychiatric Research, Orangeburg, New York, USA
| | - Eric Klann
- Center for Neural Science, New York University, New York, New York, USA
| | - Charles Hoeffer
- Institute for Behavioral Genetics, University of Colorado, Boulder, Colorado, USA
| | - Linnaea Ostroff
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut, USA
| |
Collapse
|
15
|
Fischer PD, Papadopoulos E, Dempersmier JM, Wang ZF, Nowak RP, Donovan KA, Kalabathula J, Gorgulla C, Junghanns PPM, Kabha E, Dimitrakakis N, Petrov OI, Mitsiades C, Ducho C, Gelev V, Fischer ES, Wagner G, Arthanari H. A biphenyl inhibitor of eIF4E targeting an internal binding site enables the design of cell-permeable PROTAC-degraders. Eur J Med Chem 2021; 219:113435. [PMID: 33892272 DOI: 10.1016/j.ejmech.2021.113435] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/23/2021] [Accepted: 04/01/2021] [Indexed: 12/15/2022]
Abstract
The eukaryotic translation initiation factor 4E (eIF4E) is the master regulator of cap-dependent protein synthesis. Overexpression of eIF4E is implicated in diseases such as cancer, where dysregulation of oncogenic protein translation is frequently observed. eIF4E has been an attractive target for cancer treatment. Here we report a high-resolution X-ray crystal structure of eIF4E in complex with a novel inhibitor (i4EG-BiP) that targets an internal binding site, in contrast to the previously described inhibitor, 4EGI-1, which binds to the surface. We demonstrate that i4EG-BiP is able to displace the scaffold protein eIF4G and inhibit the proliferation of cancer cells. We provide insights into how i4EG-BiP is able to inhibit cap-dependent translation by increasing the eIF4E-4E-BP1 interaction while diminishing the interaction of eIF4E with eIF4G. Leveraging structural details, we designed proteolysis targeted chimeras (PROTACs) derived from 4EGI-1 and i4EG-BiP and characterized these on biochemical and cellular levels. We were able to design PROTACs capable of binding eIF4E and successfully engaging Cereblon, which targets proteins for proteolysis. However, these initial PROTACs did not successfully stimulate degradation of eIF4E, possibly due to competitive effects from 4E-BP1 binding. Our results highlight challenges of targeted proteasomal degradation of eIF4E that must be addressed by future efforts.
Collapse
Affiliation(s)
- Patrick D Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA; Department of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University, Saarbrücken, 66123, Germany
| | - Evangelos Papadopoulos
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA; Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
| | - Jon M Dempersmier
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Zi-Fu Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Radosław P Nowak
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Katherine A Donovan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Joann Kalabathula
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Christoph Gorgulla
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Pierre P M Junghanns
- Department of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University, Saarbrücken, 66123, Germany
| | - Eihab Kabha
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Nikolaos Dimitrakakis
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Ognyan I Petrov
- Faculty of Chemistry and Pharmacy, Sofia University, 1 James Bourchier Blvd., 1164, Sofia, Bulgaria
| | | | - Christian Ducho
- Department of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University, Saarbrücken, 66123, Germany
| | - Vladimir Gelev
- Faculty of Chemistry and Pharmacy, Sofia University, 1 James Bourchier Blvd., 1164, Sofia, Bulgaria
| | - Eric S Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Gerhard Wagner
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Haribabu Arthanari
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
16
|
Devulapalli R, Jones N, Farrell K, Musaus M, Kugler H, McFadden T, Orsi SA, Martin K, Nelsen J, Navabpour S, O'Donnell M, McCoig E, Jarome TJ. Males and females differ in the regulation and engagement of, but not requirement for, protein degradation in the amygdala during fear memory formation. Neurobiol Learn Mem 2021; 180:107404. [PMID: 33609735 DOI: 10.1016/j.nlm.2021.107404] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/28/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023]
Abstract
Over the last decade, strong evidence has emerged that protein degradation mediated by the ubiquitin-proteasome system is critical for fear memory formation in the amygdala. However, this work has been done primarily in males, leaving unanswered questions about whether females also require protein degradation during fear memory formation. Here, we found that male and female rats differed in their engagement and regulation of, but not need for, protein degradation in the amygdala during fear memory formation. Male, but not female, rats had increased protein degradation in the nuclei of amygdala cells after fear conditioning. Conversely, females had elevated baseline levels of overall ubiquitin-proteasome activity in amygdala nuclei. Gene expression and DNA methylation analyses identified that females had increased baseline expression of the ubiquitin coding gene Uba52, which had increased DNA 5-hydroxymethylation (5hmc) in its promoter region, indicating a euchromatin state necessary for increased levels of ubiquitin in females. Consistent with this, persistent CRISPR-dCas9 mediated silencing of Uba52 and proteasome subunit Psmd14 in the amygdala reduced baseline protein degradation levels and impaired fear memory in male and female rats, while enhancing baseline protein degradation in the amygdala of both sexes promoted fear memory formation. These results suggest that while both males and females require protein degradation in the amygdala for fear memory formation, they differ in their baseline regulation and engagement of this process following learning. These results have important implications for understanding the etiology of sex-related differences in fear memory formation.
Collapse
Affiliation(s)
- Rishi Devulapalli
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Natalie Jones
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Kayla Farrell
- Department of Animal and Poultry Science, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Madeline Musaus
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Hannah Kugler
- Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Taylor McFadden
- Department of Animal and Poultry Science, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Sabrina A Orsi
- Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Kiley Martin
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Jacob Nelsen
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Shaghayegh Navabpour
- Fralin Biomedical Research Institute, Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA, USA
| | - Madison O'Donnell
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Emmarose McCoig
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Timothy J Jarome
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA; Department of Animal and Poultry Science, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA; Fralin Biomedical Research Institute, Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA, USA.
| |
Collapse
|
17
|
Shrestha P, Shan Z, Mamcarz M, Ruiz KSA, Zerihoun AT, Juan CY, Herrero-Vidal PM, Pelletier J, Heintz N, Klann E. Amygdala inhibitory neurons as loci for translation in emotional memories. Nature 2020; 586:407-411. [PMID: 33029009 PMCID: PMC7572709 DOI: 10.1038/s41586-020-2793-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 07/06/2020] [Indexed: 01/09/2023]
Abstract
To survive in a dynamic environment, animals need to identify and appropriately respond to stimuli that signal danger1. Survival also depends on suppressing the threat-response during a stimulus that predicts the absence of threat (safety)2-5. An understanding of the biological substrates of emotional memories during a task in which animals learn to flexibly execute defensive responses to a threat-predictive cue and a safety cue is critical for developing treatments for memory disorders such as post-traumatic stress disorder5. The centrolateral amygdala is an important node in the neuronal circuit that mediates defensive responses6-9, and a key brain area for processing and storing threat memories. Here we applied intersectional chemogenetic strategies to inhibitory neurons in the centrolateral amygdala of mice to block cell-type-specific translation programs that are sensitive to depletion of eukaryotic initiation factor 4E (eIF4E) and phosphorylation of eukaryotic initiation factor 2α (p-eIF2α). We show that de novo translation in somatostatin-expressing inhibitory neurons in the centrolateral amygdala is necessary for the long-term storage of conditioned-threat responses, whereas de novo translation in protein kinase Cδ-expressing inhibitory neurons in the centrolateral amygdala is necessary for the inhibition of a conditioned response to a safety cue. Our results provide insight into the role of de novo protein synthesis in distinct inhibitory neuron populations in the centrolateral amygdala during the consolidation of long-term memories.
Collapse
Affiliation(s)
- Prerana Shrestha
- Center for Neural Science, New York University, New York, NY, USA.
| | - Zhe Shan
- Center for Neural Science, New York University, New York, NY, USA
| | - Maggie Mamcarz
- Center for Neural Science, New York University, New York, NY, USA
| | | | - Adam T Zerihoun
- Center for Neural Science, New York University, New York, NY, USA
| | - Chien-Yu Juan
- Center for Neural Science, New York University, New York, NY, USA
| | | | - Jerry Pelletier
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Nathaniel Heintz
- Laboratory of Molecular Biology, The Rockefeller University, New York, NY, USA
| | - Eric Klann
- Center for Neural Science, New York University, New York, NY, USA.
- NYU Neuroscience Institute, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
18
|
Wang X, Chen K, Pan M, Ge W, He Z. Comparison of proteome alterations during aging in the temporal lobe of humans and rhesus macaques. Exp Brain Res 2020; 238:1963-1976. [PMID: 32572507 DOI: 10.1007/s00221-020-05855-2] [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: 02/06/2020] [Accepted: 06/11/2020] [Indexed: 01/06/2023]
Abstract
Rhesus macaques are widely used as animal models for studies of the nervous system; however, it is unknown whether the alterations in the protein profile of the brain during aging are conserved between humans and rhesus macaques. In this study, temporal cortex samples from old and young humans (84 vs. 34 years, respectively) or rhesus macaques (20 vs. 6 years, respectively) were subjected to tandem mass tag-labeled proteomic analysis followed by bioinformatic analysis. A total of 3861 homologous pairs of proteins were identified during the aging process. The conservatively upregulated proteins (n = 190) were involved mainly in extracellular matrix (ECM), focal adhesion and coagulation; while, the conservatively downregulated proteins (n = 56) were enriched in ribosome. Network analysis showed that these conservatively regulated proteins interacted with each other with respect to protein synthesis and cytoskeleton-ECM connection. Many proteins in the focal adhesion, blood clotting, complement and coagulation, and cytoplasmic ribosomal protein pathways were regulated in the same direction in human and macaque; while, proteins involved in oligodendrocyte specification and differentiation pathways were downregulated during human aging, and many proteins in the electron transport chain pathway showed differences in the altered expression profiles. Data are available via ProteomeXchange with identifier PXD013597. Our findings suggest similarities in some changes in brain protein profiles during aging both in humans and macaques, although other changes are unique to only one of these species.
Collapse
Affiliation(s)
- Xia Wang
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Kang Chen
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Meng Pan
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Wei Ge
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China. .,Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China.
| | - Zhanlong He
- Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Disease, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China.
| |
Collapse
|
19
|
Kordestani-Moghadam P, Nasehi M, Khodagholi F, Vaseghi S, Zarrindast MR, Khani M. The fluctuations of metabotropic glutamate receptor subtype 5 (mGluR5) in the amygdala in fear conditioning model of male Wistar rats following sleep deprivation, reverse circadian and napping. Brain Res 2020; 1734:146739. [PMID: 32087111 DOI: 10.1016/j.brainres.2020.146739] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/15/2020] [Accepted: 02/18/2020] [Indexed: 12/20/2022]
Abstract
Sleep is involved in metabolic system, mental health and cognitive functions. Evidence shows that sleep deprivation (SD) negatively affects mental health and impairs cognitive functions, including learning and memory. Furthermore, the metabotropic glutamate receptor subtype 5 (mGluR5) is a metabolic biomarker, which is affected by various conditions, including stress, sleep deprivation, and cognitive and psychiatric disorders. In this research, we investigated the effect of SD and reverse circadian (RC), and two models of napping (continuous and non-continuous) combined with SD or RC on fear-conditioning memory, anxiety-like behavior and mGluR5 fluctuations in the amygdala. 64 male Wistar rats were used in this study. The water box apparatus was used to induce SD/RC for 48 h, and fear-conditioning memory apparatus was used to assess fear memory. The results showed, fear-conditioning memory was impaired following SD and RC, especially in contextual stage. However, anxiety-like behavior was increased. Furthermore, mGluR5 was increased in the left amygdala more than the right amygdala. Additionally, continuous napping significantly improved fear-conditioning memory, especially freezing behavior. In conclusion, following SD and RC, fear-conditioning memory in contextual stage is more vulnerable than in auditory stage. Furthermore, increase in anxiety-like behavior is related to increase in the activity of left amygdala and mGluR5 receptors.
Collapse
Affiliation(s)
| | - Mohammad Nasehi
- Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Salar Vaseghi
- Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad-Reza Zarrindast
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Department of Neuroendocrinology, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran; Institute for Cognitive Science Studies (ICSS), Tehran, Iran
| | - Mojgan Khani
- Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
20
|
Cell-type-specific drug-inducible protein synthesis inhibition demonstrates that memory consolidation requires rapid neuronal translation. Nat Neurosci 2020; 23:281-292. [PMID: 31959934 PMCID: PMC7147976 DOI: 10.1038/s41593-019-0568-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 12/05/2019] [Indexed: 12/04/2022]
Abstract
New protein synthesis is known to be required for the consolidation of memories, yet existing methods to block translation lack spatiotemporal precision and cell-type specificity, preventing investigation of cell-specific contributions of protein synthesis. Here, we developed a combined knock-in mouse and chemogenetic approach for cell type-specific and drug-inducible protein synthesis inhibition (ciPSI) that enables rapid and reversible phosphorylation of eIF2α, leading to inhibition of general translation by 50% in vivo. We use ciPSI to show that targeted protein synthesis inhibition pan-neuronally and in excitatory neurons in lateral amygdala (LA) impaired long-term memory. This could be recovered with artificial chemogenetic activation of LA neurons, though at the cost of stimulus generalization. Conversely, genetically reducing phosphorylation of eIF2α in excitatory neurons in LA enhanced memory strength, but reduced memory fidelity and behavioral flexibility. Our findings provide evidence for a cell-specific translation program during consolidation of threat memories.
Collapse
|
21
|
Hagerty JR, Jolly ER. Heads or tails? Differential translational regulation in cercarial heads and tails of schistosome worms. PLoS One 2019; 14:e0224358. [PMID: 31658287 PMCID: PMC6816793 DOI: 10.1371/journal.pone.0224358] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 10/11/2019] [Indexed: 12/20/2022] Open
Abstract
Schistosomes are obligate helminths responsible for over 218 million cases of human schistosomiasis in 78 countries around the world. Infection occurs when free-swimming cercariae penetrate human skin and initiate developmental progression into parasitic obligate worms that consume red blood cells. Transcriptomic studies of infectious cercariae reveal abundant mRNAs associated with energy metabolism and host invasion. However, the cercaria is mostly transcriptionally quiescent, suggesting that most mRNAs are primed prior to cercarial escape from the snail host. The use of transcriptomics to understand protein expression presumes that transcription and translation are functionally coupled and the cercarial stage has categorically been treated as a single unit for -omic analysis. Per contra, the relationship between transcription and translation in infectious cercariae has not been described. To understand the correlation between transcription and translation in cercariae, we separately measured nascent translation levels in cercarial heads, cercarial tails and in the developing schistosomula, the next stage of its life cycle. The loss of the cercarial tail is essential for the transformation from a cercaria to a schistosomulum. We observed that translation was initially limited and the translation rate accelerated during the first 72-hours after tail loss. When we tested nascent translation in cercarial heads, cercarial tails, whole cercariae, and 4-hour schistosomula, we found that translation is significantly upregulated in the cercarial tail when compared to the cercarial head and that translation was undetectable in heads using immunofluorescent image quantification (p = .0005). These data represent a major shift in how we understand the cercarial stage. The cercarial head is mostly transcriptionally and translationally quiescent while being sufficient for progression into a schistosomulum. In addition, transcription and translation are not linked in Schistosoma mansoni cercaria. Thus, our current conceptual approach of treating the cercaria as a single functional unit for -omic studies may be insufficient to understand cercarial development.
Collapse
Affiliation(s)
- James R. Hagerty
- Case Western Reserve University, Department of Biology, Cleveland, OH, United States of America
| | - Emmitt R. Jolly
- Case Western Reserve University, Department of Biology, Cleveland, OH, United States of America
- Case Western Reserve University, Center for Global Health and Disease, Cleveland, OH, United States of America
- * E-mail:
| |
Collapse
|
22
|
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
|
23
|
Madan JS, Gupta K, Chattarji S, Bhattacharya A. Hippocampal and amygdalar cell-specific translation is similar soon after stress but diverge over time. Hippocampus 2019; 28:441-452. [PMID: 29626848 DOI: 10.1002/hipo.22845] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 03/16/2018] [Accepted: 03/26/2018] [Indexed: 01/16/2023]
Abstract
Stress is known to cause contrasting patterns of morphological and physiological plasticity in the hippocampus and amygdala. An obligatory cellular process underlying such neural changes is de novo translation and alterations in protein expression. Yet the nature of the translational response to stress in neurons remains largely unexplored. Even less is known about how glia are affected. Using a click-chemistry-based method to label the de novo proteome in live brain slices, we monitored translation in neurons and astrocytes of the basolateral amygdala (BLA) and dorsal hippocampal area CA3 (dCA3) in rats at different time-points after a single 2-hr exposure to immobilization stress. We observed enhancements in neuronal translation in both brain regions 1 hour after stress. This initial increase persisted in the BLA up to 10 days afterwards. In contrast, dCA3 neuronal translation gradually decreased to below control levels 10 days later. Translation profiles of dCA3 astrocytes followed timelines similar to neurons, but in BLA astrocytes translation peaked 1 day later and remained elevated 10 days later. Together our results demonstrate that stress causes an immediate upregulation of protein synthesis in both amygdalar and hippocampal neurons and astrocytes. However, these two areas eventually exhibit opposite temporal profiles of protein expression well after the end of stress. These findings identify new metrics of stress-induced plasticity at the level of cell-type specific proteomic landscape that may provide important insights into the molecular basis of the divergent temporal effects of stress across brain regions and biological scales.
Collapse
Affiliation(s)
- Jesvin S Madan
- National Centre for Biological Sciences, Tata Institute for Fundamental Research, Bangalore, 560065, India
| | - Kanika Gupta
- National Centre for Biological Sciences, Tata Institute for Fundamental Research, Bangalore, 560065, India
| | - Sumantra Chattarji
- National Centre for Biological Sciences, Tata Institute for Fundamental Research, Bangalore, 560065, India.,Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, 560065, India.,Center for Integrative Physiology, Deanery of Biomedical Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH89XD, United Kingdom
| | - Aditi Bhattacharya
- Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, 560065, India
| |
Collapse
|
24
|
Koren SA, Hamm MJ, Meier SE, Weiss BE, Nation GK, Chishti EA, Arango JP, Chen J, Zhu H, Blalock EM, Abisambra JF. Tau drives translational selectivity by interacting with ribosomal proteins. Acta Neuropathol 2019; 137:571-583. [PMID: 30759285 PMCID: PMC6426815 DOI: 10.1007/s00401-019-01970-9] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 12/23/2022]
Abstract
There is a fundamental gap in understanding the consequences of tau-ribosome interactions. Tau oligomers and filaments hinder protein synthesis in vitro, and they associate strongly with ribosomes in vivo. Here, we investigated the consequences of tau interactions with ribosomes in transgenic mice, in cells, and in human brain tissues to identify tau as a direct modulator of ribosomal selectivity. First, we performed microarrays and nascent proteomics to measure changes in protein synthesis. Using regulatable rTg4510 tau transgenic mice, we determined that tau expression differentially shifts both the transcriptome and the nascent proteome, and that the synthesis of ribosomal proteins is reversibly dependent on tau levels. We further extended these results to human brains and found that tau pathologically interacts with ribosomal protein S6 (rpS6 or S6), a crucial regulator of translation. Consequently, protein synthesis under translational control of rpS6 was reduced under tauopathic conditions in Alzheimer's disease brains. Our data establish tau as a driver of RNA translation selectivity. Moreover, since regulation of protein synthesis is critical for learning and memory, aberrant tau-ribosome interactions in disease could explain the linkage between tauopathies and cognitive impairment.
Collapse
Affiliation(s)
- Shon A Koren
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, University of Florida, 1275 Center Drive, BOX 100159, Gainesville, FL, 32610, USA
| | - Matthew J Hamm
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, University of Florida, 1275 Center Drive, BOX 100159, Gainesville, FL, 32610, USA
| | - Shelby E Meier
- Sanders Brown Center on Aging, Department of Physiology, Spinal Cord and Brain Injury Research Center, and Epilepsy Center, University of Kentucky, Lexington, KY, 40513, USA
| | - Blaine E Weiss
- Sanders Brown Center on Aging, Department of Physiology, Spinal Cord and Brain Injury Research Center, and Epilepsy Center, University of Kentucky, Lexington, KY, 40513, USA
| | - Grant K Nation
- Sanders Brown Center on Aging, Department of Physiology, Spinal Cord and Brain Injury Research Center, and Epilepsy Center, University of Kentucky, Lexington, KY, 40513, USA
| | - Emad A Chishti
- Sanders Brown Center on Aging, Department of Physiology, Spinal Cord and Brain Injury Research Center, and Epilepsy Center, University of Kentucky, Lexington, KY, 40513, USA
| | - Juan Pablo Arango
- Sanders Brown Center on Aging, Department of Physiology, Spinal Cord and Brain Injury Research Center, and Epilepsy Center, University of Kentucky, Lexington, KY, 40513, USA
| | - Jing Chen
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40513, USA
| | - Haining Zhu
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40513, USA
| | - Eric M Blalock
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, 40513, USA
| | - Jose F Abisambra
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, University of Florida, 1275 Center Drive, BOX 100159, Gainesville, FL, 32610, USA.
- Sanders Brown Center on Aging, Department of Physiology, Spinal Cord and Brain Injury Research Center, and Epilepsy Center, University of Kentucky, Lexington, KY, 40513, USA.
| |
Collapse
|
25
|
Schmitz F, Chao MV, Wyse ATS. Methylphenidate alters Akt-mTOR signaling in rat pheochromocytoma cells. Int J Dev Neurosci 2018; 73:10-18. [PMID: 30578823 DOI: 10.1016/j.ijdevneu.2018.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/13/2018] [Accepted: 12/17/2018] [Indexed: 12/12/2022] Open
Abstract
The exponential increase in methylphenidate (MPH) prescriptions in recent years has worried researchers about its misuse among individuals who do not meet the full diagnostic criteria for attention-deficit/hyperactivity disorder (ADHD) such as young children and students in search of cognitive improvement or for recreational reasons. The action of MPH is based mainly on inhibition of dopamine transporter, but the complete cellular effects are still unknown. Based upon prior studies, we attempted to determine whether the treatment with MPH (1μM) influences protein kinase B-mammalian target of rapamycin complex 1 signaling pathways (Akt-mTOR), including translation repressor protein (4E-BP1) and mitogen activated protein kinase (S6K), in rat pheochromocytoma cells (PC12), a well characterized cellular model, in a long or short term. MPH effects on the Akt substrates [cAMP response element-binding protein (CREB), forkhead box protein O1 (FoxO1), and glycogen synthase kinase 3 beta (GSK-3β)] were also evaluated. Whereas short term MPH treatment decreased the pAkt/Akt, pmTOR/mTOR and pS6K/S6K ratios, as well as pFoxO1 immunocontent in PC12 cells, long term treatment increased pAkt/Akt, pmTOR/mTOR and pGSK-3β/GSK-3β ratio. Phosphorylation levels of 4E-BP1 were decreased at 15 and 30 min and increased at 1 and 6 h by MPH. pCREB/CREB ratio was decreased. This study shows that the Akt-mTOR pathway, as well as other important Akt substrates which have been described as important regulators of protein synthesis, as well as being implicated in cellular survival, synaptic plasticity and memory consolidation, was affected by MPH in PC12 cells, representing an important step in exploring the MPH effects.
Collapse
Affiliation(s)
- Felipe Schmitz
- Departments of Cell Biology, Physiology & Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York University, New York, NY, USA; Laboratory of Neuroprotection and Metabolic Diseases, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| | - Moses V Chao
- Departments of Cell Biology, Physiology & Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York University, New York, NY, USA
| | - Angela T S Wyse
- Laboratory of Neuroprotection and Metabolic Diseases, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Programa de Pós‑Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
26
|
Uttam S, Wong C, Price TJ, Khoutorsky A. eIF4E-Dependent Translational Control: A Central Mechanism for Regulation of Pain Plasticity. Front Genet 2018; 9:470. [PMID: 30459806 PMCID: PMC6232926 DOI: 10.3389/fgene.2018.00470] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 09/24/2018] [Indexed: 01/04/2023] Open
Abstract
Translational control of gene expression has emerged as a key mechanism in regulating different forms of long-lasting neuronal plasticity. Maladaptive plastic reorganization of peripheral and spinal nociceptive circuits underlies many chronic pain states and relies on new gene expression. Accordingly, downregulation of mRNA translation in primary afferents and spinal dorsal horn neurons inhibits tissue injury-induced sensitization of nociceptive pathways, supporting a central role for translation dysregulation in the development of persistent pain. Translation is primarily regulated at the initiation stage via the coordinated activity of translation initiation factors. The mRNA cap-binding protein, eukaryotic translation initiation factor 4E (eIF4E), is involved in the recruitment of the ribosome to the mRNA cap structure, playing a central role in the regulation of translation initiation. eIF4E integrates inputs from the mTOR and ERK signaling pathways, both of which are activated in numerous painful conditions to regulate the translation of a subset of mRNAs. Many of these mRNAs are involved in the control of cell growth, proliferation, and neuroplasticity. However, the full repertoire of eIF4E-dependent mRNAs in the nervous system and their translation regulatory mechanisms remain largely unknown. In this review, we summarize the current evidence for the role of eIF4E-dependent translational control in the sensitization of pain circuits and present pharmacological approaches to target these mechanisms. Understanding eIF4E-dependent translational control mechanisms and their roles in aberrant plasticity of nociceptive circuits might reveal novel therapeutic targets to treat persistent pain states.
Collapse
Affiliation(s)
- Sonali Uttam
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Calvin Wong
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Theodore J. Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, United States
- Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX, United States
| | - Arkady Khoutorsky
- Department of Anesthesia, McGill University, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
| |
Collapse
|
27
|
Nguyen PV, Gelinas JN. Noradrenergic gating of long-lasting synaptic potentiation in the hippocampus: from neurobiology to translational biomedicine. J Neurogenet 2018; 32:171-182. [DOI: 10.1080/01677063.2018.1497630] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Peter V. Nguyen
- Department of Physiology and Institute of Neuroscience & Mental Health, University of Alberta School of Medicine, Edmonton, Canada
| | - Jennifer N. Gelinas
- Department of Neurology and Institute for Genomic Medicine, College of Physicians & Surgeons of Columbia University, New York, NY,USA
| |
Collapse
|
28
|
Chu J, Pelletier J. Therapeutic Opportunities in Eukaryotic Translation. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a032995. [PMID: 29440069 DOI: 10.1101/cshperspect.a032995] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The ability to block biological processes with selective small molecules provides advantages distinct from most other experimental approaches. These include rapid time to onset, swift reversibility, ability to probe activities in manners that cannot be accessed by genetic means, and the potential to be further developed as therapeutic agents. Small molecule inhibitors can also be used to alter expression and activity without affecting the stoichiometry of interacting partners. These tenets have been especially evident in the field of translation. Small molecule inhibitors were instrumental in enabling investigators to capture short-lived complexes and characterize specific steps of protein synthesis. In addition, several drugs that are the mainstay of modern antimicrobial drug therapy are potent inhibitors of prokaryotic translation. Currently, there is much interest in targeting eukaryotic translation as decades of research have revealed that deregulated protein synthesis in cancer cells represents a targetable vulnerability. In addition to being potential therapeutics, small molecules that manipulate translation have also been shown to influence cognitive processes such as memory. In this review, we focus on small molecule modulators that target the eukaryotic translation initiation apparatus and provide an update on their potential application to the treatment of disease.
Collapse
Affiliation(s)
- Jennifer Chu
- Department of Biochemistry, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Jerry Pelletier
- Department of Biochemistry, McGill University, Montreal, Quebec H3G 1Y6, Canada.,Department of Oncology, McGill University, Montreal, Quebec H3G 1Y6, Canada.,Rosalind and Morris Goodman Cancer Research Center, McGill University, Montreal, Quebec H3G 1Y6, Canada
| |
Collapse
|
29
|
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
|
30
|
Ravi V, Jain A, Ahamed F, Fathma N, Desingu PA, Sundaresan NR. Systematic evaluation of the adaptability of the non-radioactive SUnSET assay to measure cardiac protein synthesis. Sci Rep 2018; 8:4587. [PMID: 29545554 PMCID: PMC5854694 DOI: 10.1038/s41598-018-22903-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 03/01/2018] [Indexed: 11/09/2022] Open
Abstract
Heart is a dynamic organ that undergoes remodeling in response to both physiological and pathological stimuli. One of the fundamental cellular processes that facilitates changes in the size and shape of this muscular organ is the protein synthesis. Traditionally changes in cardiac protein synthesis levels were measured by radiolabeled tracers. However, these methods are often cumbersome and suffer from radioactive risk. Recently a nonradioactive method for detecting protein synthesis under in vitro conditions called the Surface Sensing of Translation (SUnSET) was described in cell lines of mouse dendrites and T cells. In this work, we provide multiple lines of evidence that the SUnSET assay can be applied to reliably detect changes in protein synthesis both in isolated neonatal primary cardiomyocytes and heart. We successfully tracked the changes in protein synthesis by western blotting as well as immunohistochemical variants of the SUnSET assay. Applying the SUnSET assay, we measured the cardiac protein synthesis during the different ages of mice. Further, we successfully tracked the increase in cardiac protein synthesis during different stages of a well-established model for pathological hypertrophy. Overall, we propose SUnSET assay as a simple, reliable and robust method to measure protein synthesis in the cardiac milieu.
Collapse
Affiliation(s)
- Venkatraman Ravi
- Cardiovascular and Muscle Research Laboratory, Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| | - Aditi Jain
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| | - Faiz Ahamed
- Cardiovascular and Muscle Research Laboratory, Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| | - Nowrin Fathma
- Cardiovascular and Muscle Research Laboratory, Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| | - Perumal Arumugam Desingu
- Cardiovascular and Muscle Research Laboratory, Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| | - Nagalingam R Sundaresan
- Cardiovascular and Muscle Research Laboratory, Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India. .,Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India.
| |
Collapse
|
31
|
Rajmohan R, Reddy PH. Amyloid-Beta and Phosphorylated Tau Accumulations Cause Abnormalities at Synapses of Alzheimer's disease Neurons. J Alzheimers Dis 2018; 57:975-999. [PMID: 27567878 DOI: 10.3233/jad-160612] [Citation(s) in RCA: 349] [Impact Index Per Article: 49.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Amyloid-beta (Aβ) and hyperphosphorylated tau are hallmark lesions of Alzheimer's disease (AD). However, the loss of synapses and dysfunctions of neurotransmission are more directly tied to disease severity. The role of these lesions in the pathoetiological progression of the disease remains contested. Biochemical, cellular, molecular, and pathological studies provided several lines of evidence and improved our understanding of how Aβ and hyperphosphorylated tau accumulation may directly harm synapses and alter neurotransmission. In vitro evidence suggests that Aβ and hyperphosphorylated tau have both direct and indirect cytotoxic effects that affect neurotransmission, axonal transport, signaling cascades, organelle function, and immune response in ways that lead to synaptic loss and dysfunctions in neurotransmitter release. Observations in preclinical models and autopsy studies support these findings, suggesting that while the pathoetiology of positive lesions remains elusive, their removal may reduce disease severity and progression. The purpose of this article is to highlight the need for further investigation of the role of tau in disease progression and its interactions with Aβ and neurotransmitters alike.
Collapse
Affiliation(s)
- Ravi Rajmohan
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Cell Biology & Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Speech, Language and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
32
|
Psychedelics and reconsolidation of traumatic and appetitive maladaptive memories: focus on cannabinoids and ketamine. Psychopharmacology (Berl) 2018; 235:433-445. [PMID: 29178010 DOI: 10.1007/s00213-017-4793-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 11/14/2017] [Indexed: 12/22/2022]
Abstract
RATIONALE Clinical data with 3,4-methylenedioxymethamphetamine (MDMA) in post-traumatic stress disorder (PTSD) patients recently stimulated interest on the potential therapeutic use of psychedelics in disorders characterized by maladaptive memories, including substance use disorders (SUD). The rationale for the use of MDMA in PTSD and SUD is being extended to a broader beneficial "psychedelic effect," which is supporting further clinical investigations, in spite of the lack of mechanistic hypothesis. Considering that the retrieval of emotional memories reactivates specific brain mechanisms vulnerable to inhibition, interference, or strengthening (i.e., the reconsolidation process), it was proposed that the ability to retrieve and change these maladaptive memories might be a novel intervention for PTSD and SUD. The mechanisms underlying MDMA effects indicate memory reconsolidation modulation as a hypothetical process underlying its efficacy. OBJECTIVE Mechanistic and clinical studies with other two classes of psychedelic substances, namely cannabinoids and ketamine, are providing data in support of a potential use in PTSD and SUD based on the modulation of traumatic and appetitive memory reconsolidation, respectively. Here, we review preclinical and clinical data on cannabinoids and ketamine effects on biobehavioral processes related to the reconsolidation of maladaptive memories. RESULTS We report the findings supporting (or not) the working hypothesis linking the potential therapeutic effect of these substances to the underlying reconsolidation process. We also proposed possible approaches for testing the use of these two classes of drugs within the current paradigm of reconsolidation memory inhibition. CONCLUSIONS Metaplasticity may be the process in common between cannabinoids and ketamine/ketamine-like substance effects on the mediation and potential manipulation of maladaptive memories.
Collapse
|
33
|
Fattore L, Piva A, Zanda MT, Fumagalli G, Chiamulera C. Psychedelics and reconsolidation of traumatic and appetitive maladaptive memories: focus on cannabinoids and ketamine. Psychopharmacology (Berl) 2017. [PMID: 29178010 DOI: 10.1007/s00213-017-4793-4.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/29/2022]
Abstract
RATIONALE Clinical data with 3,4-methylenedioxymethamphetamine (MDMA) in post-traumatic stress disorder (PTSD) patients recently stimulated interest on the potential therapeutic use of psychedelics in disorders characterized by maladaptive memories, including substance use disorders (SUD). The rationale for the use of MDMA in PTSD and SUD is being extended to a broader beneficial "psychedelic effect," which is supporting further clinical investigations, in spite of the lack of mechanistic hypothesis. Considering that the retrieval of emotional memories reactivates specific brain mechanisms vulnerable to inhibition, interference, or strengthening (i.e., the reconsolidation process), it was proposed that the ability to retrieve and change these maladaptive memories might be a novel intervention for PTSD and SUD. The mechanisms underlying MDMA effects indicate memory reconsolidation modulation as a hypothetical process underlying its efficacy. OBJECTIVE Mechanistic and clinical studies with other two classes of psychedelic substances, namely cannabinoids and ketamine, are providing data in support of a potential use in PTSD and SUD based on the modulation of traumatic and appetitive memory reconsolidation, respectively. Here, we review preclinical and clinical data on cannabinoids and ketamine effects on biobehavioral processes related to the reconsolidation of maladaptive memories. RESULTS We report the findings supporting (or not) the working hypothesis linking the potential therapeutic effect of these substances to the underlying reconsolidation process. We also proposed possible approaches for testing the use of these two classes of drugs within the current paradigm of reconsolidation memory inhibition. CONCLUSIONS Metaplasticity may be the process in common between cannabinoids and ketamine/ketamine-like substance effects on the mediation and potential manipulation of maladaptive memories.
Collapse
Affiliation(s)
- Liana Fattore
- National Research Council of Italy, Institute of Neuroscience-Cagliari, Cagliari, Italy
| | - Alessandro Piva
- Sezione Farmacologia, Dipt. Diagnostica e Sanità Pubblica, Università degli Studi di Verona, Policlinico Borgo Roma, P.le Scuro 10, 37134, Verona, Italy
| | - Mary Tresa Zanda
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Cittadella Universitaria di Monserrato, SP 8, Km 0.700, 09042, Monserrato, Italy
| | - Guido Fumagalli
- Sezione Farmacologia, Dipt. Diagnostica e Sanità Pubblica, Università degli Studi di Verona, Policlinico Borgo Roma, P.le Scuro 10, 37134, Verona, Italy
| | - Cristiano Chiamulera
- Sezione Farmacologia, Dipt. Diagnostica e Sanità Pubblica, Università degli Studi di Verona, Policlinico Borgo Roma, P.le Scuro 10, 37134, Verona, Italy.
| |
Collapse
|
34
|
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
|
35
|
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]
|
36
|
Heise C, Taha E, Murru L, Ponzoni L, Cattaneo A, Guarnieri FC, Montani C, Mossa A, Vezzoli E, Ippolito G, Zapata J, Barrera I, Ryazanov AG, Cook J, Poe M, Stephen MR, Kopanitsa M, Benfante R, Rusconi F, Braida D, Francolini M, Proud CG, Valtorta F, Passafaro M, Sala M, Bachi A, Verpelli C, Rosenblum K, Sala C. eEF2K/eEF2 Pathway Controls the Excitation/Inhibition Balance and Susceptibility to Epileptic Seizures. Cereb Cortex 2017; 27:2226-2248. [PMID: 27005990 PMCID: PMC5963824 DOI: 10.1093/cercor/bhw075] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Alterations in the balance of inhibitory and excitatory synaptic transmission have been implicated in the pathogenesis of neurological disorders such as epilepsy. Eukaryotic elongation factor 2 kinase (eEF2K) is a highly regulated, ubiquitous kinase involved in the control of protein translation. Here, we show that eEF2K activity negatively regulates GABAergic synaptic transmission. Indeed, loss of eEF2K increases GABAergic synaptic transmission by upregulating the presynaptic protein Synapsin 2b and α5-containing GABAA receptors and thus interferes with the excitation/inhibition balance. This cellular phenotype is accompanied by an increased resistance to epilepsy and an impairment of only a specific hippocampal-dependent fear conditioning. From a clinical perspective, our results identify eEF2K as a potential novel target for antiepileptic drugs, since pharmacological and genetic inhibition of eEF2K can revert the epileptic phenotype in a mouse model of human epilepsy.
Collapse
Affiliation(s)
| | - Elham Taha
- Sagol Department of Neurobiology and
- Center for Gene Manipulation in the Brain, Natural Science Faculty, University of Haifa, Haifa, Israel
| | - Luca Murru
- CNR Neuroscience Institute, Milan, Italy
| | - Luisa Ponzoni
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | | | - Fabrizia C. Guarnieri
- Division of Neuroscience, San Raffaele Scientific Institute and Vita-Salute University, Milan, Italy
| | | | | | - Elena Vezzoli
- CNR Neuroscience Institute, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | | | | | - Iliana Barrera
- Sagol Department of Neurobiology and
- Center for Gene Manipulation in the Brain, Natural Science Faculty, University of Haifa, Haifa, Israel
| | - Alexey G. Ryazanov
- The Department of Pharmacology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - James Cook
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Michael Poe
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Michael Rajesh Stephen
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Maksym Kopanitsa
- Synome, Babraham Research Campus, Cambridge CB22 3AT, UK
- Charles River Discovery Research Services, 70210 Kuopio, Finland
| | - Roberta Benfante
- CNR Neuroscience Institute, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Francesco Rusconi
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Daniela Braida
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Maura Francolini
- CNR Neuroscience Institute, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Christopher G. Proud
- University of Southampton, Centre for Biological Sciences, Southampton SO17 1BJ, UK
- South Australian Health and Medical Research Institute and University of Adelaide, Adelaide, Australia
| | - Flavia Valtorta
- Division of Neuroscience, San Raffaele Scientific Institute and Vita-Salute University, Milan, Italy
| | - Maria Passafaro
- CNR Neuroscience Institute, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Mariaelvina Sala
- CNR Neuroscience Institute, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Angela Bachi
- IFOM-FIRC Institute of Molecular Oncology, Milan, Italy
| | - Chiara Verpelli
- CNR Neuroscience Institute, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Kobi Rosenblum
- Sagol Department of Neurobiology and
- Center for Gene Manipulation in the Brain, Natural Science Faculty, University of Haifa, Haifa, Israel
| | - Carlo Sala
- CNR Neuroscience Institute, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
37
|
Rizzo V, Touzani K, Raveendra BL, Swarnkar S, Lora J, Kadakkuzha BM, Liu XA, Zhang C, Betel D, Stackman RW, Puthanveettil SV. Encoding of contextual fear memory requires de novo proteins in the prelimbic cortex. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2017; 2:158-169. [PMID: 28503670 DOI: 10.1016/j.bpsc.2016.10.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Despite our understanding of the significance of the prefrontal cortex in the consolidation of long-term memories (LTM), its role in the encoding of LTM remains elusive. Here we investigated the role of new protein synthesis in the mouse medial prefrontal cortex (mPFC) in encoding contextual fear memory. METHODS Because a change in the association of mRNAs to polyribosomes is an indicator of new protein synthesis, we assessed the changes in polyribosome-associated mRNAs in the mPFC following contextual fear conditioning (CFC) in the mouse. Differential gene expression in mPFC was identified by polyribosome profiling (n = 18). The role of new protein synthesis in mPFC was determined by focal inhibition of protein synthesis (n = 131) and by intra-prelimbic cortex manipulation (n = 56) of Homer 3, a candidate identified from polyribosome profiling. RESULTS We identified several mRNAs that are differentially and temporally recruited to polyribosomes in the mPFC following CFC. Inhibition of protein synthesis in the prelimbic (PL), but not in the anterior cingulate cortex (ACC) region of the mPFC immediately after CFC disrupted encoding of contextual fear memory. Intriguingly, inhibition of new protein synthesis in the PL 6 hours after CFC did not impair encoding. Furthermore, expression of Homer 3, an mRNA enriched in polyribosomes following CFC, in the PL constrained encoding of contextual fear memory. CONCLUSIONS Our studies identify several molecular substrates of new protein synthesis in the mPFC and establish that encoding of contextual fear memories require new protein synthesis in PL subregion of mPFC.
Collapse
Affiliation(s)
- Valerio Rizzo
- Department of Neuroscience, The Scripps Research Institute, Scripps Florida 130 Scripps Way, Jupiter, FL 33458
| | - Khalid Touzani
- Department of Neuroscience, The Scripps Research Institute, Scripps Florida 130 Scripps Way, Jupiter, FL 33458
| | - Bindu L Raveendra
- Department of Neuroscience, The Scripps Research Institute, Scripps Florida 130 Scripps Way, Jupiter, FL 33458
| | - Supriya Swarnkar
- Department of Neuroscience, The Scripps Research Institute, Scripps Florida 130 Scripps Way, Jupiter, FL 33458
| | - Joan Lora
- Department of Psychology, Center for Complex Systems & Brain Sciences, College of Science, Florida Atlantic University, Jupiter, FL 33458
| | - Beena M Kadakkuzha
- Department of Neuroscience, The Scripps Research Institute, Scripps Florida 130 Scripps Way, Jupiter, FL 33458
| | - Xin-An Liu
- Department of Neuroscience, The Scripps Research Institute, Scripps Florida 130 Scripps Way, Jupiter, FL 33458
| | - Chao Zhang
- Department of Medicine and Institute for Computational Biomedicine, Weill Cornell Medical College, New York. NY10065. USA
| | - Doron Betel
- Department of Medicine and Institute for Computational Biomedicine, Weill Cornell Medical College, New York. NY10065. USA
| | - Robert W Stackman
- Department of Psychology, Center for Complex Systems & Brain Sciences, College of Science, Florida Atlantic University, Jupiter, FL 33458
| | | |
Collapse
|
38
|
Accumulation of Polyribosomes in Dendritic Spine Heads, But Not Bases and Necks, during Memory Consolidation Depends on Cap-Dependent Translation Initiation. J Neurosci 2017; 37:1862-1872. [PMID: 28087764 DOI: 10.1523/jneurosci.3301-16.2017] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/09/2016] [Accepted: 01/09/2017] [Indexed: 12/28/2022] Open
Abstract
Translation in dendrites is believed to support synaptic changes during memory consolidation. Although translational control mechanisms are fundamental mediators of memory, little is known about their role in local translation. We previously found that polyribosomes accumulate in dendritic spines of the adult rat lateral amygdala (LA) during consolidation of aversive pavlovian conditioning and that this memory requires cap-dependent initiation, a primary point of translational control in eukaryotic cells. Here we used serial electron microscopy reconstructions to quantify polyribosomes in LA dendrites when consolidation was blocked by the cap-dependent initiation inhibitor 4EGI-1. We found that 4EGI-1 depleted polyribosomes in dendritic shafts and selectively prevented their upregulation in spine heads, but not bases and necks, during consolidation. Cap-independent upregulation was specific to spines with small, astrocyte-associated synapses. Our results reveal that cap-dependent initiation is involved in local translation during learning and that local translational control varies with synapse type.SIGNIFICANCE STATEMENT Translation initiation is a central regulator of long-term memory formation. Local translation in dendrites supports memory by providing necessary proteins at synaptic sites, but it is unknown whether this requires initiation or bypasses it. We used serial electron microscopy reconstructions to examine polyribosomes in dendrites when memory formation was blocked by an inhibitor of translation initiation. This revealed two major pools of polyribosomes that were upregulated during memory formation: one pool in dendritic spine heads that was initiation dependent and another pool in the bases and necks of small spines that was initiation independent. Thus, translation regulation differs between spine types and locations, and translation that occurs closest to individual synapses during memory formation is initiation dependent.
Collapse
|
39
|
Hussein NA, Delaney TL, Tounsel BL, Liebl FLW. The Extracellular-Regulated Kinase Effector Lk6 is Required for Glutamate Receptor Localization at the Drosophila Neuromuscular Junction. J Exp Neurosci 2016; 10:77-91. [PMID: 27199570 PMCID: PMC4866800 DOI: 10.4137/jen.s32840] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 03/14/2016] [Accepted: 03/18/2016] [Indexed: 11/16/2022] Open
Abstract
The proper localization and synthesis of postsynaptic glutamate receptors are essential for synaptic plasticity. Synaptic translation initiation is thought to occur via the target of rapamycin (TOR) and mitogen-activated protein kinase signal-integrating kinase (Mnk) signaling pathways, which is downstream of extracellular-regulated kinase (ERK). We used the model glutamatergic synapse, the Drosophila neuromuscular junction, to better understand the roles of the Mnk and TOR signaling pathways in synapse development. These synapses contain non-NMDA receptors that are most similar to AMPA receptors. Our data show that Lk6, the Drosophila homolog of Mnk1 and Mnk2, is required in either presynaptic neurons or postsynaptic muscle for the proper localization of the GluRIIA glutamate receptor subunit. Lk6 may signal through eukaryotic initiation factor (eIF) 4E to regulate the synaptic levels of GluRIIA as either interfering with eIF4E binding to eIF4G or expression of a nonphosphorylatable isoform of eIF4E resulted in a significant reduction in GluRIIA at the synapse. We also find that Lk6 and TOR may independently regulate synaptic levels of GluRIIA.
Collapse
Affiliation(s)
- Nizar A Hussein
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| | - Taylor L Delaney
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| | - Brittany L Tounsel
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| | - Faith L W Liebl
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| |
Collapse
|
40
|
Abstract
The mammalian target of rapamycin (mTOR) is a central regulator of a diverse array of cellular processes, including cell growth, proliferation, autophagy, translation, and actin polymerization. Components of the mTOR cascade are present at synapses and influence synaptic plasticity and spine morphogenesis. A prevailing view is that the study of mTOR and its role in autism spectrum disorders (ASDs) will elucidate the molecular mechanisms by which mTOR regulates neuronal function under physiological and pathological conditions. Although many ASDs arise as a result of mutations in genes with multiple molecular functions, they appear to converge on common biological pathways that give rise to autism-relevant behaviors. Dysregulation of mTOR signaling has been identified as a phenotypic feature common to fragile X syndrome, tuberous sclerosis complex 1 and 2, neurofibromatosis 1, phosphatase and tensin homolog, and potentially Rett syndrome. Below are a summary of topics covered in a symposium that presents dysregulation of mTOR as a unifying theme in a subset of ASDs.
Collapse
|
41
|
CaMKII regulates proteasome phosphorylation and activity and promotes memory destabilization following retrieval. Neurobiol Learn Mem 2016; 128:103-9. [PMID: 26779588 DOI: 10.1016/j.nlm.2016.01.001] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 12/30/2015] [Accepted: 01/05/2016] [Indexed: 01/04/2023]
Abstract
Numerous studies have suggested that memories "destabilize" and require de novo protein synthesis in order to reconsolidate following retrieval, but very little is known about how this destabilization process is regulated. Recently, ubiquitin-proteasome mediated protein degradation has been identified as a critical regulator of memory trace destabilization following retrieval, though the specific mechanisms controlling retrieval-induced changes in ubiquitin-proteasome activity remain equivocal. Here, we found that proteasome activity is increased in the amygdala in a CaMKII-dependent manner following the retrieval of a contextual fear memory. We show that in vitro inhibition of CaMKII reversed retrieval-induced increases in proteasome activity. Additionally, in vivo pharmacological blockade of CaMKII abolished increases in proteolytic activity and activity related regulatory phosphorylation in the amygdala following retrieval, suggesting that CaMKII was "upstream" of protein degradation during the memory reconsolidation process. Consistent with this, while inhibiting CaMKII in the amygdala did not impair memory following retrieval, it completely attenuated the memory impairments that resulted from post-retrieval protein synthesis blockade. Collectively, these results suggest that CaMKII controls the initiation of the memory reconsolidation process through regulation of the proteasome.
Collapse
|
42
|
Borovok N, Nesher E, Levin Y, Reichenstein M, Pinhasov A, Michaelevski I. Dynamics of Hippocampal Protein Expression During Long-term Spatial Memory Formation. Mol Cell Proteomics 2015; 15:523-41. [PMID: 26598641 DOI: 10.1074/mcp.m115.051318] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Indexed: 01/08/2023] Open
Abstract
Spatial memory depends on the hippocampus, which is particularly vulnerable to aging. This vulnerability has implications for the impairment of navigation capacities in older people, who may show a marked drop in performance of spatial tasks with advancing age. Contemporary understanding of long-term memory formation relies on molecular mechanisms underlying long-term synaptic plasticity. With memory acquisition, activity-dependent changes occurring in synapses initiate multiple signal transduction pathways enhancing protein turnover. This enhancement facilitates de novo synthesis of plasticity related proteins, crucial factors for establishing persistent long-term synaptic plasticity and forming memory engrams. Extensive studies have been performed to elucidate molecular mechanisms of memory traces formation; however, the identity of plasticity related proteins is still evasive. In this study, we investigated protein turnover in mouse hippocampus during long-term spatial memory formation using the reference memory version of radial arm maze (RAM) paradigm. We identified 1592 proteins, which exhibited a complex picture of expression changes during spatial memory formation. Variable linear decomposition reduced significantly data dimensionality and enriched three principal factors responsible for variance of memory-related protein levels at (1) the initial phase of memory acquisition (165 proteins), (2) during the steep learning improvement (148 proteins), and (3) the final phase of the learning curve (123 proteins). Gene ontology and signaling pathways analysis revealed a clear correlation between memory improvement and learning phase-curbed expression profiles of proteins belonging to specific functional categories. We found differential enrichment of (1) neurotrophic factors signaling pathways, proteins regulating synaptic transmission, and actin microfilament during the first day of the learning curve; (2) transcription and translation machinery, protein trafficking, enhancement of metabolic activity, and Wnt signaling pathway during the steep phase of memory formation; and (3) cytoskeleton organization proteins. Taken together, this study clearly demonstrates dynamic assembly and disassembly of protein-protein interaction networks depending on the stage of memory formation engrams.
Collapse
Affiliation(s)
- Natalia Borovok
- From the ‡Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Elimelech Nesher
- §Department of Molecular Biology, Ariel University, Ariel 4070000, Israel
| | - Yishai Levin
- ¶de Botton Institute for Protein Profiling, The Nancy & Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Michal Reichenstein
- From the ‡Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Albert Pinhasov
- §Department of Molecular Biology, Ariel University, Ariel 4070000, Israel
| | - Izhak Michaelevski
- From the ‡Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel-Aviv 6997801, Israel; ‖Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
43
|
Wan J, Shi F, Xu Z, Zhao M. Knockdown of eIF4E suppresses cell proliferation, invasion and enhances cisplatin cytotoxicity in human ovarian cancer cells. Int J Oncol 2015; 47:2217-25. [PMID: 26498997 DOI: 10.3892/ijo.2015.3201] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/17/2015] [Indexed: 11/06/2022] Open
Abstract
Eukaryotic initiation factor 4E (eIF4E) plays an important role in cap-dependent translation. The overexpression of eIF4E gene has been found in a variety of human malignancies. In this study, we attempted to identify the potential effects of eIF4E and explore the possibility of eIF4E as a therapeutic target for the treatment of human ovarian cancer. First the activation of eIF4E protein was detected with m7-GTP cap binding assays in ovarian cancer and control cells. Next, the eIF4E-shRNA expression plasmids were used to specifically inhibit eIF4E activity in ovarian cancer cells line A2780 and C200. The effects of knockdown eIF4E gene on cell proliferation, migration and invasion were investigated in vitro. Moreover, the changes of cell cycle and apoptosis of ovarian cancer cells were detected by flow cytometry. Finally, we investigated the effect of knockdown of eIF4E on the chemosensitivity of ovarian cancer cells to cisplatin in vitro. Our results show there is elevated activation of eIF4E in ovarian cancer cells compared with normal human ovarian epithelial cell line. The results of BrdU incorporation and FCM assay indicate that knockdown of eIF4E efficiently suppressed cell growth and induce cell cycle arrest in G1 phase and subsequent apoptosis in ovarian cancer cells. From Transwell assay analysis, knockdown eIF4E significantly decrease cellular migration and invasion of ovarian cancer cells. We also confirmed that knockdown eIF4E could synergistically enhance the cytotoxicity effects of cisplatin to cancer cells and sensitized cisplatin-resistant C200 cells in vitro. This study demonstrates that the activation of eIF4E gene is an essential component of the malignant phenotype in ovarian cancer, and aberration of eIF4E expression is associated with proliferation, migration, invasion and chemosensitivity to cisplatin in ovarian cancer cells. Knockdown eIF4E gene can be used as a potential therapeutic target for the treatment of human ovarian cancer.
Collapse
Affiliation(s)
- Jing Wan
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Hubei 430071, P.R. China
| | - Fang Shi
- Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Hubei 430071, P.R. China
| | - Zhanzhan Xu
- Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Hubei 430071, P.R. China
| | - Min Zhao
- Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Hubei 430071, P.R. China
| |
Collapse
|
44
|
Martin I, Abalde-Atristain L, Kim JW, Dawson TM, Dawson VL. Abberant protein synthesis in G2019S LRRK2 Drosophila Parkinson disease-related phenotypes. Fly (Austin) 2015; 8:165-9. [PMID: 25483009 DOI: 10.4161/19336934.2014.983382] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
LRRK2 mutations are a frequent cause of familial Parkinson disease (PD) and are also found in a number of sporadic PD cases. PD-linked G2019S and I2020T mutations in the kinase domain of LRRK2 result in elevated kinase activity, which is required for the toxicity of these pathogenic variants in cell and animal models of PD. We recently reported that LRRK2 interacts with and phosphorylates a number of mammalian ribosomal proteins, several of which exhibit increased phosphorylation via both G2019S and I2020T LRRK2. Blocking the phosphorylation of ribosomal protein s15 through expression of phospho-deficient T136A s15 prevents age-associated locomotor deficits and dopamine neuron loss caused by G2019S LRRK2 expression in Drosophila indicating that s15 is a pathogenic LRRK2 substrate. We previously described that G2019S LRRK2 causes an induction of bulk mRNA translation that is blocked by T136A s15 or the protein synthesis inhibitor anisomycin. Here, we report the protective effects of the eIF4E/eIF4G interaction inhibitor 4EGI-1, in preventing neurodegenerative phenotypes in G2019S LRRK2 flies, and discuss how our findings and those of other groups provide a framework to begin investigating the mechanistic impact of LRRK2 on translation.
Collapse
Affiliation(s)
- Ian Martin
- a Neuroregeneration and Stem Cell Programs; Institute for Cell Engineering; Johns Hopkins University School of Medicine ; Baltimore , MD USA
| | | | | | | | | |
Collapse
|
45
|
Wang H, Huang F, Wang J, Wang P, Lv W, Hong L, Li S, Zhou J. The synergistic inhibition of breast cancer proliferation by combined treatment with 4EGI-1 and MK2206. Cell Cycle 2015; 14:232-42. [PMID: 25607647 DOI: 10.4161/15384101.2014.977096] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cap-dependent translation is a potential cancer-related target (oncotarget) due to its critical role in cancer initiation and progression. 4EGI-1, an inhibitor of eIF4E/eIF4G interaction, was discovered by screening chemical libraries of small molecules. 4EGI-1 inhibits cap-dependent translation initiation by impairing the assembly of the eIF4E/eIF4G complex, and therefore is a potential anti-cancer agent. Here, we report that 4EGI-1 also inhibits mTORC1 signaling independent of its inhibitory role on cap-dependent translation initiation. The inhibition of mTORC1 signaling by 4EGI-1 activates Akt due to both abrogation of the negative feedback loops from mTORC1 to PI3K and activation of mTORC2. We further validated that mTORC2 activity is required for 4EGI-1-mediated Akt activation. The activated Akt counteracted the anticancer effects of 4EGI-1. In support of this model, inhibition of Akt potentiates the antitumor activity of 4EGI-1 both in vitro and in a xenograft mouse model in vivo. Our results suggest that a combination of 4EGI-1and Akt inhibitor is a rational approach for the treatment of cancer.
Collapse
Affiliation(s)
- Hongtao Wang
- a Laboratory of Medical Molecular Biology; Beijing Institute of Biotechnology ; Beijing , P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Maity S, Rah S, Sonenberg N, Gkogkas CG, Nguyen PV. Norepinephrine triggers metaplasticity of LTP by increasing translation of specific mRNAs. ACTA ACUST UNITED AC 2015; 22:499-508. [PMID: 26373828 PMCID: PMC4579357 DOI: 10.1101/lm.039222.115] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 06/30/2015] [Indexed: 12/24/2022]
Abstract
Norepinephrine (NE) is a key modulator of synaptic plasticity in the hippocampus, a brain structure crucially involved in memory formation. NE boosts synaptic plasticity mostly through initiation of signaling cascades downstream from beta (β)-adrenergic receptors (β-ARs). Previous studies demonstrated that a β-adrenergic receptor agonist, isoproterenol, can modify the threshold for long-term potentiation (LTP), a putative cellular mechanism for learning and memory, in a process known as “metaplasticity.” Metaplasticity is the ability of synaptic plasticity to be modified by prior experience. We asked whether NE itself could engage metaplastic mechanisms in area CA1 of mouse hippocampal slices. Using extracellular field potential recording and stimulation, we show that application of NE (10 µM), which did not alter basal synaptic strength, enhances the future maintenance of LTP elicited by subthreshold, high-frequency stimulation (HFS: 1 × 100 Hz, 1 sec). HFS applied 30 min after NE washout induced long-lasting (>4 h) LTP, which was significantly extended in duration relative to HFS alone. This NE-induced metaplasticity required β1-AR activation, as coapplication of the β1-receptor antagonist CGP-20712A (1 µM) attenuated maintenance of LTP. We also found that NE-mediated metaplasticity was translation- and transcription-dependent. Polysomal profiles of CA1 revealed increased translation rates for specific mRNAs during NE-induced metaplasticity. Thus, activation of β-ARs by NE primes synapses for future long-lasting plasticity on time scales extending beyond fast synaptic transmission; this may facilitate neural information processing and the subsequent formation of lasting memories.
Collapse
Affiliation(s)
- Sabyasachi Maity
- Department of Physiology, University of Alberta School of Medicine, Edmonton, Alberta T6G 2H7, Canada
| | - Sean Rah
- Department of Physiology, University of Alberta School of Medicine, Edmonton, Alberta T6G 2H7, Canada
| | - Nahum Sonenberg
- Department of Biochemistry, Goodman Cancer Centre, McGill University, Montreal, Quebec H3A 1A3, Canada
| | - Christos G Gkogkas
- Patrick Wild Centre and Centre for Integrative Biology, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| | - Peter V Nguyen
- Department of Physiology, University of Alberta School of Medicine, Edmonton, Alberta T6G 2H7, Canada Department of Psychiatry, University of Alberta School of Medicine, Edmonton, Alberta T6G 2H7, Canada Neuroscience and Mental Health Institute, University of Alberta School of Medicine, Edmonton, Alberta T6G 2H7, Canada
| |
Collapse
|
47
|
O'Dell TJ, Connor SA, Guglietta R, Nguyen PV. β-Adrenergic receptor signaling and modulation of long-term potentiation in the mammalian hippocampus. ACTA ACUST UNITED AC 2015; 22:461-71. [PMID: 26286656 PMCID: PMC4561407 DOI: 10.1101/lm.031088.113] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/02/2015] [Indexed: 02/06/2023]
Abstract
Encoding new information in the brain requires changes in synaptic strength. Neuromodulatory transmitters can facilitate synaptic plasticity by modifying the actions and expression of specific signaling cascades, transmitter receptors and their associated signaling complexes, genes, and effector proteins. One critical neuromodulator in the mammalian brain is norepinephrine (NE), which regulates multiple brain functions such as attention, perception, arousal, sleep, learning, and memory. The mammalian hippocampus receives noradrenergic innervation and hippocampal neurons express β-adrenergic receptors, which are known to play important roles in gating the induction of long-lasting forms of synaptic potentiation. These forms of long-term potentiation (LTP) are believed to importantly contribute to long-term storage of spatial and contextual memories in the brain. In this review, we highlight the contributions of noradrenergic signaling in general and β-adrenergic receptors in particular, toward modulating hippocampal LTP. We focus on the roles of NE and β-adrenergic receptors in altering the efficacies of specific signaling molecules such as NMDA and AMPA receptors, protein phosphatases, and translation initiation factors. Also, the roles of β-adrenergic receptors in regulating synaptic "tagging" and "capture" of LTP within synaptic networks of the hippocampus are reviewed. Understanding the molecular and cellular bases of noradrenergic signaling will enrich our grasp of how the brain makes new, enduring memories, and may shed light on credible strategies for improving mental health through treatment of specific disorders linked to perturbed memory processing and dysfunctional noradrenergic synaptic transmission.
Collapse
Affiliation(s)
- Thomas J O'Dell
- Department of Physiology, David Geffen School of Medicine and Integrative Center for Learning and Memory, Brain Research Institute, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Steven A Connor
- Department of Physiology, University of Alberta School of Medicine, Edmonton, Alberta T6G 2H7, Canada
| | - Ryan Guglietta
- Interdepartmental Ph.D. Program for Neuroscience, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Peter V Nguyen
- Department of Physiology, University of Alberta School of Medicine, Edmonton, Alberta T6G 2H7, Canada Department of Psychiatry, University of Alberta School of Medicine, Edmonton, Alberta T6G 2H7, Canada Department of Neuroscience & Mental Health Institute, University of Alberta School of Medicine, Edmonton, Alberta T6G 2H7, Canada
| |
Collapse
|
48
|
Baldi E, Bucherelli C. Brain sites involved in fear memory reconsolidation and extinction of rodents. Neurosci Biobehav Rev 2015; 53:160-90. [DOI: 10.1016/j.neubiorev.2015.04.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 03/30/2015] [Accepted: 04/06/2015] [Indexed: 12/21/2022]
|
49
|
Buffington SA, Huang W, Costa-Mattioli M. Translational control in synaptic plasticity and cognitive dysfunction. Annu Rev Neurosci 2015; 37:17-38. [PMID: 25032491 DOI: 10.1146/annurev-neuro-071013-014100] [Citation(s) in RCA: 267] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Activity-dependent changes in the strength of synaptic connections are fundamental to the formation and maintenance of memory. The mechanisms underlying persistent changes in synaptic strength in the hippocampus, specifically long-term potentiation and depression, depend on new protein synthesis. Such changes are thought to be orchestrated by engaging the signaling pathways that regulate mRNA translation in neurons. In this review, we discuss the key regulatory pathways that govern translational control in response to synaptic activity and the mRNA populations that are specifically targeted by these pathways. The critical contribution of regulatory control over new protein synthesis to proper cognitive function is underscored by human disorders associated with either silencing or mutation of genes encoding proteins that directly regulate translation. In light of these clinical implications, we also consider the therapeutic potential of targeting dysregulated translational control to treat cognitive disorders of synaptic dysfunction.
Collapse
Affiliation(s)
- Shelly A Buffington
- Department of Neuroscience, Memory and Brain Research Center, Baylor College of Medicine, Houston, Texas 77030; , ,
| | | | | |
Collapse
|
50
|
Luyck K, Luyten L. Can electrical stimulation of the human bed nucleus of the stria terminalis reduce contextual anxiety? An unanswered question. Front Behav Neurosci 2015; 9:69. [PMID: 25852509 PMCID: PMC4362315 DOI: 10.3389/fnbeh.2015.00069] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 03/01/2015] [Indexed: 11/13/2022] Open
Affiliation(s)
- Kelly Luyck
- Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, KU Leuven Leuven, Belgium
| | - Laura Luyten
- Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, KU Leuven Leuven, Belgium ; Psychology of Learning and Experimental Psychopathology, Psychology and Educational Sciences, KU Leuven Leuven, Belgium
| |
Collapse
|