Translational control by eIF2α kinases in long-lasting synaptic plasticity and long-term memory

https://doi.org/10.1016/j.nlm.2013.04.013Get rights and content

Highlights

  • eIF2α kinases control general and gene-specific translation.

  • eIF2α kinases are required for hippocampal synaptic plasticity.

  • eIF2α kinases are required for various forms of long-term memory.

Abstract

Although the requirement for new protein synthesis in synaptic plasticity and memory has been well established, recent genetic, molecular, electrophysiological, and pharmacological studies have broadened our understanding of the translational control mechanisms that are involved in these processes. One of the critical translational control points mediating general and gene-specific translation depends on the phosphorylation of eukaryotic initiation factor 2 alpha (eIF2α) by four regulatory kinases. Here, we review the literature highlighting the important role for proper translational control via regulation of eIF2α phosphorylation by its kinases in long-lasting synaptic plasticity and long-term memory.

Introduction

One of the more remarkable features of the brain is the ability to acquire and store new information as lasting memory traces. This continuous capacity to learn and remember allows one to process changes in the environment, retain new information, and adapt to behavioral choices over time. A fundamental question remains that intrigues modern neuroscientists: how are memories formed and stored at the cellular and molecular level? Behavioral studies performed in mice treated with the protein synthesis inhibitor puromycin provided the first molecular clue that protein synthesis is required for long-term memory (LTM) formation, but not for task acquisition and short-term memory (STM) formation (Flexner, Flexner, & Stellar, 1963). Since then, a plethora of pharmacological and genetic studies have highlighted the critical role for de novo gene expression and protein synthesis in LTM formation (Kandel, 2001, McGaugh, 2000).

Neurons can alter their molecular and physiological characteristics in response to temporal- and activity-dependent changes in their environment. Synaptic plasticity refers to the ability of the brain to change the efficacy (strengthening or weakening) of synaptic connections between neurons and is hypothesized as the cellular basis for learning and memory (Bliss and Collingridge, 1993, Malenka and Nicoll, 1999). These persistent, activity-dependent, changes in synaptic strength are triggered by de novo protein synthesis (Klann & Sweatt, 2008). Evidence indicating a role for protein synthesis at local synaptic sites stem from observations that neuronal dendrites and their spines contain polyribosomes (Steward & Levy, 1982), translation factors (Tang & Schuman, 2002), and mRNA (Crino & Eberwine, 1996) that can be translated into proteins to support synaptic activity. Consistent with this notion, local protein synthesis was shown to be necessary for long-lasting increases in synaptic strength induced by brain-derived neurotrophic factor (BDNF; Kang & Schuman, 1996). Similarly, rapid, local protein synthesis also was required for long-lasting decreases in synaptic strength induced by activation of group I metabotropic glutamate receptors (mGluR; Huber, Kayser, & Bear, 2000). Together, these findings indicate that protein synthesis can be triggered locally at activated synapses and is required for persistent, activity-dependent forms of synaptic plasticity, which in turn is thought to be essential for memory formation.

Although the initial report from Flexner et al. (1963) and other early studies identified new protein synthesis as a molecular requirement for memory formation, they offered little in the way of molecular translational control mechanisms because they relied mostly on the administration of general translation inhibitors into animals. In the last 10 years, however, a vast amount of genetic, biochemical, pharmacological, and physiological studies have increased our knowledge of the precise translational control mechanisms underlying long-lasting synaptic plasticity, memory formation, and cognitive function (Costa-Mattioli et al., 2009, Kelleher et al., 2004, Richter and Klann, 2009). In this review, we specifically discuss the functional role of eIF2α kinases and their regulation of activity-dependent synaptic plasticity and cognitive function, including learning and memory.

Section snippets

Translational control by eIF2α phosphorylation

Translational control can be defined as a change in either the efficiency or rate of mRNA translation. The process of mRNA translation can be divided into three main steps: initiation, elongation, and termination. Although regulation can occur at each step, translational control primarily occurs at the rate-limiting initiation step when the small 40S ribosomal subunit is recruited to the mRNA and positioned at the initiation codon (Jackson, Hellen, & Pestova, 2010). Translation initiation

eIF2α kinases

eIF2α phosphorylation is regulated by four serine/threonine (Ser/Thr) protein kinases, each of which phosphorylate eIF2α on Ser51. The four eIF2α kinases are heme-regulated inhibitor (HRI), the double-stranded (ds) RNA activated protein kinase (PKR), the general control non-derepressible-2 (GCN2), and the PKR-like endoplasmic reticulum (ER) resident protein kinase (PERK). These four eIF2α kinases share a conserved kinase domain, but respond differentially to various cellular stressors due to

GCN2 controls L-LTP and LTM

As previously mentioned, distinct eIF2α kinases phosphorylate eIF2α to control two fundamental processes that are crucial for the consolidation of long-term memories: de novo protein synthesis and CREB-mediated gene expression via the memory repressing factor ATF4. Evidence that regulation of eIF2α phosphorylation plays an important role in long-lasting synaptic plasticity and LTM was first provided by the observation that GCN2-deficient mice exhibit a lowered threshold for the induction of

PKR controls gene-specific translation, L-LTP, and LTM

Using a novel pharmacogenetic mouse model, Jiang et al. (2010) demonstrated that a selective increase in PKR-mediated eIF2α phosphorylation in CA1 hippocampal neurons impairs L-LTP and LTM. Accordingly, the increase in eIF2α phosphorylation was associated with an increase in ATF4 translation and the suppression of CREB-dependent gene expression (Jiang et al., 2010), including that of BDNF, a key protein involved in L-LTP and the consolidation of LTM (Bekinschtein et al., 2008). It should be

PERK controls ATF4 translation and behavioral flexibility

Using mouse behavioral genetics and biochemistry, it was demonstrated that PERK plays a crucial role in regulating behavioral flexibility (Trinh et al., 2012). Previous studies showed that global inactivation of PERK results in severe developmental abnormalities (Wei et al., 2008, Zhang et al., 2002). To rule out the possibility that the effects of PERK on synaptic and cognitive function occur during development, mice were generated in which PERK was selectively removed in the forebrain at

Conclusions and future directions

It is now evident that eIF2α kinases and their ability to impact translational control via the phosphorylation of eIF2α are critical for long-lasting plasticity of synaptic connections and for cognitive functions that rely on such plasticity in the brain. One intriguing insight from the studies of the role of eIF2α kinases in synaptic plasticity thus far is that eIF2α phosphorylation is divergently regulated during LTP and LTD. Moreover, these studies suggest that eIF2α kinases, such as PERK

References (98)

  • M. Costa-Mattioli et al.

    EIF2alpha phosphorylation bidirectionally regulates the switch from short- to long-term synaptic plasticity and memory

    Cell

    (2007)
  • M. Costa-Mattioli et al.

    Translational control of long-lasting synaptic plasticity and memory

    Neuron

    (2009)
  • J. Couturier et al.

    Interaction of double-stranded RNA-dependent protein kinase (PKR) with the death receptor signaling pathway in amyloid beta (Abeta)-treated cells and in APPSLPS1 knock-in mice

    Journal of Biological Chemistry

    (2010)
  • P.B. Crino et al.

    Molecular characterization of the dendritic growth cone: Regulated mRNA transport and local protein synthesis

    Neuron

    (1996)
  • J. Deng et al.

    Activation of GCN2 in UV-irradiated cells inhibits translation

    Current Biology

    (2002)
  • S. Dey et al.

    Both transcriptional regulation and translational control of ATF4 are central to the integrated stress response

    Journal of Biological Chemistry

    (2010)
  • H.P. Harding et al.

    Regulated translation initiation controls stress-induced gene expression in mammalian cells

    Molecular Cell

    (2000)
  • H.P. Harding et al.

    Diabetes mellitus and exocrine pancreatic dysfunction in perk−/− mice reveals a role for translational control in secretory cell survival

    Molecular Cell

    (2001)
  • M. Iacono et al.

    UAUG and uORFs in human and rodent 5’untranslated mRNAs

    Gene

    (2005)
  • R.J. Kelleher et al.

    Translational regulatory mechanisms in persistent forms of synaptic plasticity

    Neuron

    (2004)
  • E. Klann et al.

    Altered protein synthesis is a trigger for long-term memory formation

    Neurobiology of Learning and Memory

    (2008)
  • Y. Ma et al.

    Delineation of a negative feedback regulatory loop that controls protein translation during endoplasmic reticulum stress

    Journal of Biological Chemistry

    (2003)
  • M. Matsui et al.

    Bioinformatic analysis of post-transcriptional regulation by uORF in human and mouse

    FEBS Letters

    (2007)
  • H. Mellor et al.

    Cloning and characterization of cDNA encoding rat hemin-sensitive initiation factor-2 alpha (eIF-2 alpha) kinase. Evidence for multitissue expression

    Journal of Biological Chemistry

    (1994)
  • E. Meurs et al.

    Molecular cloning and characterization of the human double-stranded RNA-activated protein kinase induced by interferon

    Cell

    (1990)
  • U. Ozcan et al.

    Loss of the tuberous sclerosis complex tumor suppressors triggers the unfolded protein response to regulate insulin signaling and apoptosis

    Molecular Cell

    (2008)
  • A.L. Peel et al.

    Activation of the cell stress kinase PKR in Alzheimer’s disease and human amyloid precursor protein transgenic mice

    Neurobiology of Diseases

    (2003)
  • C.M. Pereira et al.

    IMPACT, a protein preferentially expressed in the mouse brain, binds GCN1 and inhibits GCN2 activation

    Journal of Biological Chemistry

    (2005)
  • J.F. Raven et al.

    PKR and PKR-like endoplasmic reticulum kinase induce the proteasome-dependent degradation of cyclin D1 via a mechanism requiring eukaryotic initiation factor 2alpha phosphorylation

    Journal of Biological Chemistry

    (2008)
  • Y. Segev et al.

    ApoE epsilon4 is associated with eIF2alpha phosphorylation and impaired learning in young mice

    Neurobiology of Aging

    (2013)
  • M. Solomon et al.

    The neural substrates of cognitive control deficits in autism spectrum disorders

    Neuropsychologia

    (2009)
  • N. Sonenberg et al.

    Eukaryotic translation initiation factors and regulators

    Current Opinion in Structural Biology

    (2003)
  • M.A. Trinh et al.

    Brain-specific disruption of the eIF2alpha kinase PERK decreases ATF4 expression and impairs behavioral flexibility

    Cell Reports

    (2012)
  • W. Zhang et al.

    PERK EIF2AK3 control of pancreatic beta cell differentiation and proliferation is required for postnatal glucose homeostasis

    Cell Metabolism

    (2006)
  • F. Zhang et al.

    Binding of double-stranded RNA to protein kinase PKR is required for dimerization and promotes critical autophosphorylation events in the activation loop

    Journal of Biological Chemistry

    (2001)
  • P.J. Zhu et al.

    Suppression of PKR promotes network excitability and enhanced cognition by interferon-gamma-mediated disinhibition

    Cell

    (2011)
  • T. Abel et al.

    Memory suppressor genes: Inhibitory constraints on the storage of long-term memory

    Science

    (1998)
  • J.L. Banko et al.

    Regulation of eukaryotic initiation factor 4E by converging signaling pathways during metabotropic glutamate receptor-dependent long-term depression

    Journal of Neuroscience

    (2006)
  • J.L. Banko et al.

    The translation repressor 4E-BP2 is critical for eIF4F complex formation, synaptic plasticity, and memory in the hippocampus

    Journal of Neuroscience

    (2005)
  • P. Bekinschtein et al.

    BDNF is essential to promote persistence of long-term memory storage

    Proceedings of the National Academy of Sciences of the United States of America

    (2008)
  • J.J. Berlanga et al.

    Characterization of a mammalian homolog of the GCN2 eukaryotic initiation factor 2alpha kinase

    European Journal of Biochemistry

    (1999)
  • A. Bertolotti et al.

    Dynamic interaction of BiP and ER stress transducers in the unfolded-protein response

    Nature Cell Biology

    (2000)
  • T.V. Bliss et al.

    A synaptic model of memory: Long-term potentiation in the hippocampus

    Nature

    (1993)
  • M. Boyce et al.

    A selective inhibitor of eIF2alpha dephosphorylation protects cells from ER stress

    Science

    (2005)
  • C.F. Calkhoven et al.

    Translational control of C/EBPalpha and C/EBPbeta isoform expression

    Genes & Development

    (2000)
  • L.S. Carnevalli et al.

    Phosphorylation of the alpha subunit of translation initiation factor-2 by PKR mediates protein synthesis inhibition in the mouse brain during status epilepticus

    Biochemical Journal

    (2006)
  • S. Cohen-Chalamish et al.

    Dynamic refolding of IFN-gamma mRNA enables it to function as PKR activator and translation template

    Nature Chemical Biology

    (2009)
  • J.H. Connor et al.

    Growth arrest and DNA damage-inducible protein GADD34 assembles a novel signaling complex containing protein phosphatase 1 and inhibitor 1

    Molecular and Cellular Biology

    (2001)
  • M. Costa-Mattioli et al.

    Translational control of hippocampal synaptic plasticity and memory by the eIF2alpha kinase GCN2

    Nature

    (2005)
  • Cited by (0)

    View full text