Elsevier

Cell Calcium

Volume 49, Issue 5, May 2011, Pages 296-305
Cell Calcium

Review
Activity-dependent calcium signaling and ERK-MAP kinases in neurons: A link to structural plasticity of the nucleus and gene transcription regulation

https://doi.org/10.1016/j.ceca.2010.11.009Get rights and content

Abstract

Activity-dependent gene expression is important for the formation and maturation of neuronal networks, neuronal survival and for plastic modifications within mature networks. At the level of individual neurons, expression of new protein is required for dendritic branching, synapse formation and elimination. Experience-driven synaptic activity induces membrane depolarization, which in turn evokes intracellular calcium transients that are decoded according to their source and strength by intracellular calcium sensing proteins. In order to activate the gene transcription machinery of the cell, calcium signals have to be conveyed from the site of their generation in the cytoplasm to the cell nucleus. This can occur via a variety of mechanisms and with different kinetics depending on the source and amplitude of calcium influx. One mechanism involves the propagation of calcium itself, leading to nuclear calcium transients that subsequently activate transcription. The mitogen-activated protein kinase (MAPK) cascade represents a second central signaling module that transduces information from the site of calcium signal generation at the plasma membrane to the nucleus. Nuclear signaling of the MAPK cascades catalyzes the phosphorylation of transcription factors but also regulates gene transcription more globally at the level of chromatin remodeling as well as through its recently identified role in the modulation of nuclear shape. Here we discuss the possible mechanisms by which the MAPKs ERK1 and ERK2, activated by synaptically evoked calcium influx, can signal to the nucleus and regulate gene transcription. Moreover, we describe how MAPK-dependent structural plasticity of the nuclear envelope enhances nuclear calcium signaling and suggest possible implications for the regulation of gene transcription in the context of nuclear geometry.

Introduction

Individual neurons are born within the first weeks of life and persist throughout the lifetime of the animal, which can for some species exceed 100 years. During this time the cell has to undergo many experience-driven adaptations, which are reflected by remodeling of the dendritic tree, axonal growth and formation and elimination of synapses [1], [2], [3], [4], [5], [6], [7]. Such processes are controlled by electrical activity and require transcription of appropriate sets of genes in the nucleus [7], [8]. The major intracellular messenger that transduces electrical activity into gene expression is calcium [9], [10]. During development, the maturation of excitatory synapses depends on calcium influx through synaptic N-methyl-d-aspartate receptors (NMDARs) which leads to local modifications such as insertion of alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) into the spine and rearrangement of the post synaptic density (PSD) [11], [12]. Moreover, activity-dependent calcium-influx through NMDARs and voltage-gated calcium channels (VGCCs) is required to induce gene transcription events in the nucleus, which underlie widespread loss or pruning of dendritic spines and large-scale reorganization of the dendritic tree [13], [14].

LTP and LTD are considered cellular correlates of learning and memory and involve rapid and persistent remodeling of synaptic connections. Rapid processes such as AMPAR insertion/removal, remodeling of the actin cytoskeleton and spine turnover are regulated by local calcium and its downstream kinases and are thought to be independent of gene transcription and nuclear calcium signaling [15]. Long-lasting adaptations however, require synthesis of new proteins that alter neuronal function and stabilize synapses at a longer time scale [7], [8], [16], [17]. In order to decode the requirements of the synapse at the level of gene transcription, reliable delivery of the information about synaptic activity to the nucleus is essential. In principle, two strategies exist by which calcium can transform electrical activity into a nuclear gene transcription event: (1) calcium activates signaling molecules in the cytoplasm which carry the information to the nucleus via direct nuclear translocation or indirectly via signaling cascades, or (2) calcium itself crosses the nuclear envelope and acts directly within the cell nucleus.

In recent years many different calcium-dependent signaling pathways to the nucleus and their role in encoding the source and kinetics of intracellular calcium transients have been described [7], [18], [19], [20], [21]. Since calcium-dependent activation of gene-transcription eventually requires the physical translocation of a signal from the cytoplasm across the nuclear envelope it is important to consider the accessibility of the nucleus for such signaling molecules. Small molecules such as calcium or calcium bound to calmodulin may enter the nucleus unhindered via diffusion through nuclear pore complexes [22], [23], [24], [25]. However, larger molecules such as the calcium/calmodulin dependent kinases (CaMKs), protein kinase A (PKA) or the extracellular signal-regulated kinases 1 and 2 (ERK1/2) exceed the size limit for passive diffusion through the pore and thus require a mechanism that facilitates their nuclear translocation [26], [27], [28], [29]. In the first part of this review we will consider the principal mechanisms by which calcium signals can be conveyed to the nucleus focusing on nuclear entry of calcium and on nuclear signaling of the ERK1/2-MAPK cascade.

In addition to direct activation of transcription factors recent evidence suggests that ERK1/2 can regulate chromatin remodeling – a higher-order mechanism of transcriptional regulation. ERK1/2-dependent phosphorylation of histone H3 and subsequent gene transcription have been described in many cell types and more recently these processes have been demonstrated to be important for synaptic plasticity and memory formation in different areas of the brain. Finally, recent evidence suggests that calcium and ERK1/2 act in concert at the level of structural remodeling of the nuclear envelope itself. Synaptic activity initiates deep infoldings of the nuclear membrane leading to a compartmentalized nucleoplasm, an increased surface area and amplified nuclear calcium signaling. In the second part of this review, we will summarize the current knowledge about neuronal ERK1/2 signaling and discuss the new nuclear functions of ERK1/2 in more detail. Moreover, we will provide suggestions how structural plasticity of the nucleus can impact on nuclear calcium signaling and activity-dependent gene transcription.

Section snippets

Nuclear signaling mediated by calcium

Several different transcription factors and their upstream kinases are regulated by calcium [18]. However, transcription is often not directly controlled by calcium ions but requires the involvement of specific signaling molecules and adaptors, which are activated by calcium inside or outside the nucleus. Thus, several routes exist by which calcium can relay a message to the nuclear transcription machinery. For example, the presence of nuclear calcium has been demonstrated as a requirement for

Direct nuclear calcium signaling – pathways and mechanisms

How does calcium reach the nucleus? In the case of synaptic plasticity, activation of a few synapses is sufficient to induce persistent long-term potentiation through calcium-induced gene transcription. In a typical neuron these activated synapses can be widely distributed across the dendritic branches with distances of several hundreds of micrometers from the cell soma, which contains the nucleus. This raises the question how perinuclear calcium reaches sufficiently high concentrations to

Indirect nuclear calcium signaling – the ERK1/2-MAPK cascade

Apart from direct delivery of calcium itself to the nucleus, cytoplasmic calcium transients generate nuclear signals indirectly via protein kinase cascades. Several of those complex calcium-activated signaling systems coexist in neurons. We will narrow our focus on the ERK1/2-MAPK cascade because of its central and universal importance in synaptic plasticity and memory formation in many species, brain areas and types of synapses. Moreover, recent discoveries that ascribe new functions to

Nuclear translocation of ERK1/2

Understanding the trafficking and nuclear translocation of ERK1/2 in neurons from synapses to the nucleus is particularly important in the light of its physiological relevance for plasticity and learning. However, despite a large body of evidence for ERK1/2 functions in neuronal signal integration and propagation a unified model describing the mechanisms that mediate trafficking and nuclear translocation of ERK1/2 in neurons does not exist.

In order to phosphorylate transcription factors, ERK1/2

Plastic changes in nuclear geometry

Facilitation of calcium signaling within specialized compartments of neurons is essential to neuronal plasticity. One of the best known examples is the dendritic spine which can generate highly localized, supralinear calcium transients that lead to activation of many intracellular signaling pathways [130], [131], [132]. Due to its geometry the spine can integrate electrical signals very efficiently. Moreover, strong depolarization of the spine can induce morphological changes in the spine head

Closing remarks

Regulation of activity-dependent gene expression in neurons is a complex process occurring at many different levels in the cell ranging from local expression of a few proteins up to global expression of hundreds of genes. With ongoing research, new pathways are revealed by which the neuron may adequately control its protein composition. For example, the discovery that microRNAs are essential for post transcriptional regulation of genes involved in a large number of physiological processes adds

Conflict of interest statement

There is no conflict of interest.

References (152)

  • B.E. Lonze et al.

    Function and regulation of CREB family transcription factors in the nervous system

    Neuron

    (2002)
  • M.J. Berridge

    Neuronal calcium signaling

    Neuron

    (1998)
  • C.R. Rose et al.

    Stores not just for storage. intracellular calcium release and synaptic plasticity

    Neuron

    (2001)
  • J.D. Sweatt

    Mitogen-activated protein kinases in synaptic plasticity and memory

    Curr. Opin. Neurobiol.

    (2004)
  • J.M. Schmitt et al.

    Calcium activation of ERK mediated by calmodulin kinase I

    J. Biol. Chem.

    (2004)
  • H.J. Chen et al.

    A synaptic Ras-GTPase activating protein (p135 SynGAP) inhibited by CaM kinase II

    Neuron

    (1998)
  • S.C. Shalin et al.

    Kinase suppressor of Ras1 compartmentalizes hippocampal signal transduction and subserves synaptic plasticity and memory formation

    Neuron

    (2006)
  • M.J. Kim et al.

    Differential roles of NR2A- and NR2B-containing NMDA receptors in Ras-ERK signaling and AMPA receptor trafficking

    Neuron

    (2005)
  • J.J. Zhu et al.

    Ras and Rap control AMPA receptor trafficking during synaptic plasticity

    Cell

    (2002)
  • J. Kim et al.

    Regulation of dendritic excitability by activity-dependent trafficking of the A-type K+ channel subunit Kv4.2 in hippocampal neurons

    Neuron

    (2007)
  • R. Treisman

    The serum response element

    Trends Biochem. Sci.

    (1992)
  • B. Derijard et al.

    JNK1: a protein kinase stimulated by UV light and Ha-Ras that binds and phosphorylates the c-Jun activation domain

    Cell

    (1994)
  • E. Borrelli et al.

    Decoding the epigenetic language of neuronal plasticity

    Neuron

    (2008)
  • M. Maletic-Savatic et al.

    Rapid dendritic morphogenesis in CA1 hippocampal dendrites induced by synaptic activity

    Science

    (1999)
  • W.C. Sin et al.

    Dendrite growth increased by visual activity requires NMDA receptor and Rho GTPases

    Nature

    (2002)
  • L.C. Katz et al.

    Synaptic activity and the construction of cortical circuits

    Science

    (1996)
  • T.N. Wiesel et al.

    Single-cell responses in striate cortex of kittens deprived of vision in one eye

    J. Neurophysiol.

    (1963)
  • M. Verhage et al.

    Synaptic assembly of the brain in the absence of neurotransmitter secretion

    Science

    (2000)
  • S. Cohen et al.

    Communication between the synapse and the nucleus in neuronal development, plasticity, and disease

    Annu. Rev. Cell Dev. Biol.

    (2008)
  • S.W. Flavell et al.

    Signaling mechanisms linking neuronal activity to gene expression and plasticity of the nervous system

    Annu. Rev. Neurosci.

    (2008)
  • H. Bading

    Transcription-dependent neuronal plasticity: the nuclear calcium hypothesis

    Eur. J. Biochem.

    (2000)
  • G.E. Hardingham et al.

    Distinct functions of nuclear and cytoplasmic calcium in the control of gene expression

    Nature

    (1997)
  • G. Wu et al.

    Maturation of a central glutamatergic synapse

    Science

    (1996)
  • I. Ehrlich et al.

    Postsynaptic density 95 controls AMPA receptor incorporation during long-term potentiation and experience-driven synaptic plasticity

    J. Neurosci.

    (2004)
  • L.S. Lerea et al.

    NMDA and non-NMDA receptor-mediated increase of c-fos mRNA in dentate gyrus neurons involves calcium influx via different routes

    J. Neurosci.

    (1992)
  • H. Bading et al.

    Regulation of gene expression in hippocampal neurons by distinct calcium signaling pathways

    Science

    (1993)
  • V.A. Derkach et al.

    Regulatory mechanisms of AMPA receptors in synaptic plasticity

    Nat. Rev. Neurosci.

    (2007)
  • P.V. Nguyen et al.

    Requirement of a critical period of transcription for induction of a late phase of LTP

    Science

    (1994)
  • F.H. Cruzalegui et al.

    Calcium-regulated protein kinase cascades and their transcription factor targets

    Cell Mol. Life Sci.

    (2000)
  • A.E. West et al.

    Regulation of transcription factors by neuronal activity

    Nat. Rev. Neurosci.

    (2002)
  • P. Lipp et al.

    Nuclear calcium signalling by individual cytoplasmic calcium puffs

    EMBO J.

    (1997)
  • O. Gerasimenko et al.

    New aspects of nuclear calcium signalling

    J. Cell Sci.

    (2004)
  • A. Eder et al.

    Calcium signals can freely cross the nuclear envelope in hippocampal neurons: somatic calcium increases generate nuclear calcium transients

    BMC Neurosci.

    (2007)
  • M.D. Bootman et al.

    An update on nuclear calcium signalling

    J. Cell Sci.

    (2009)
  • R. Peters

    Translocation through the nuclear pore complex: selectivity and speed by reduction-of-dimensionality

    Traffic

    (2005)
  • P.L. Paine et al.

    Nuclear envelope permeability

    Nature

    (1975)
  • S. Chawla et al.

    CBP: a signal-regulated transcriptional coactivator controlled by nuclear calcium and CaM kinase IV

    Science

    (1998)
  • F. Ledo et al.

    Ca2+-dependent block of CREB-CBP transcription by repressor DREAM

    EMBO J.

    (2002)
  • B. Mellstrom et al.

    Ca2+-operated transcriptional networks: molecular mechanisms and in vivo models

    Physiol. Rev.

    (2008)
  • F. Shibasaki et al.

    Role of kinases and the phosphatase calcineurin in the nuclear shuttling of transcription factor NF-AT4

    Nature

    (1996)
  • Cited by (0)

    View full text