Elsevier

Brain Research

Volume 1379, 16 March 2011, Pages 44-52
Brain Research

Research Report
LIM kinase mediates estrogen action on the actin depolymerization factor Cofilin

https://doi.org/10.1016/j.brainres.2010.07.067Get rights and content

Abstract

The ovarian hormone estrogen increases the axospinous synapse density in the hippocampal CA1 region of young female rats but fails to do so in aged rats. This estrogen-mediated alteration of spine synapse structures suggests the coincident requirement for the structural reorganization of the underlying actin cytoskeleton network. Actin reorganization is known to require the deactivation of Cofilin, an actin depolymerization factor. Cofilin is deactivated by LIM kinase (LIMK), and LIMK activity is modulated by the phosphorylation of specific residues. We have previously demonstrated that estrogen is able to increase phosphorylated LIMK (pLIMK) immunoreactivity (IR) in the hippocampus in vivo and that this estrogen-stimulated pLIMK-IR is decreased in the aged brain. Because Cofilin phosphorylation allows for actin filament elongation and spine synapse growth, we sought to determine if estrogen acts through Cofilin and if such estrogen action requires the observed LIMK activity. Using both hippocampal neurons and the NG108-15 neuroblastoma cell line, we demonstrate here that estrogen stimulates the phosphorylation of Cofilin in vitro. Furthermore, this estrogen action on Cofilin requires LIMK. Lastly, while initiating the phosphorylation of LIMK and Cofilin, estrogen can also stimulate the formation of filopodial extensions, an early step in the formation of nascent spines, demonstrating that estrogen can alter the actin-dependent neuronal morphology. This linkage of estrogen communication to Cofilin via LIMK provides the functionality to the age-sensitive pLIMK-IR that we have observed in vivo.

Research Highlights

►Estrogen can alter actin-dependent morphology. ►Estrogen stimulates the phosphorylation of LIM Kinase (LIMK). ►Estrogen stimulates the phosphorylation of Cofilin. ►Estrogen action on Cofilin is LIMK-dependent.

Introduction

The hippocampal formation remains receptive and responsive to experience and to stimuli throughout adulthood. This responsiveness includes a sensitivity to circulating steroid hormones that results in the morphological remodeling of hippocampal neurons in vivo (McEwen, 2002). Across the estrous cycle of adult female rats, hippocampal CA1 pyramidal neurons respond to cyclical increases in the circulating estrogen levels with corresponding increases in dendritic spine density and axospinous synapse number (McEwen and Milner, 2007, Woolley and McEwen, 1992). The observation of such estrogen-induced spinogenesis in vivo has been reiterated in vitro with early video microscopy studies on cultured primary hippocampal neurons where stimulation with supra-physiological levels of estrogen induces the formation of immature spines, or neurite filopodia (Brinton, 1993). Together, these in vivo and in vitro observations of estrogen-modulated morphologic plasticity suggest that estrogen directly communicates with and affects the structural reorganization of the cytoskeletal network in hippocampal neurons (Dillon and Goda, 2005, Matus, 2000, Schubert and Dotti, 2007). However, this ability of estrogen to modulate dendritic spine plasticity is lost in the aged female brain (Adams and Morrison, 2003, Morrison et al., 2006).

In the aged female brain, estrogen fails to increase CA1 spine synapse density, presumably due to a loss of the required molecular communication from estrogen to the underlying actin cytoskeletal network (Yildirim et al., 2008). Actin filaments, the major cytoskeletal protein structures in the dendritic spine, affect synaptic plasticity by regulating the formation of synaptic structures as well as by affecting dendritic spine size and shape (Cingolani and Goda, 2008, Sekino et al., 2007). Estrogen may be involved in regulating spine synapse formation by regulating the actin cytoskeletal network. In support of this possibility, we have recently observed that estrogen can increase phosphorylated (p) LIM Kinase (LIMK) immunoreactivity (IR) in the hippocampus in vivo (Spencer et al., 2008a), and that this estrogen-stimulated pLIMK-IR in neurons is diminished in the aged brain (Yildirim et al., 2008).

LIMK is a signal transduction intermediate that is essential for spine formation and synapse function (Sarmiere and Bamburg, 2002) and plays a key role in regulating actin polymerization and depolymerization (Bernard, 2007). LIMK functions as a serine/threonine kinase, and one LIMK target substrate of interest is Cofilin (Yang et al., 1998). Cofilin, a member of the actin depolymerization factor (ADF) family, is a key actin binding protein that promotes the turnover and severing of actin filaments (Bernstein and Bamburg, 2010, Maciver and Hussey, 2002). Cofilin plays an important role in neuronal shape (Sarmiere and Bamburg, 2004), and Cofilin is localized to neuronal structures with high actin filament turnover, including the growth cone of axons and dendrites (Fass et al., 2004, Gungabissoon and Bamburg, 2003). Importantly, Cofilin is present in CA1 spines and is enriched near the membrane and in the postsynaptic density (PSD) where it can regulate actin dynamics in the context of synapse activity and plasticity (Racz and Weinberg, 2006). LIMK phosphorylates Cofilin, rendering Cofilin severing inactive and thereby promoting actin filament extension and dendritic spine formation (Arber et al., 1998, Endo et al., 2007, Meng et al., 2003).

To identify a functional role for the estrogen-stimulated pLIMK-IR that is observed in vivo, we asked whether estrogen affects the phosphorylation of the LIMK substrate Cofilin in cultured hippocampal neurons and in NG108-15 neuronal cells in vitro. We determined that estrogen can stimulate the phosphorylation of both Cofilin and LIMK in vitro, and we demonstrate here that the phospho-modulation of Cofilin by estrogen is LIMK-dependent.

Section snippets

Estrogen stimulates the phosphorylation of ADF/Cofilin

Cofilin is an actin-binding protein that regulates the actin assembly into microfilaments (Bernstein and Bamburg, 2010, Maciver and Hussey, 2002). Cofilin promotes the severing of filamentous actin (F-actin) that results in an increased total number of shorter actin filaments and actin monomers (Kiuchi et al., 2007). Cofilin activity is regulated by the phosphorylation of the amino-terminal Ser3 residue whereupon Cofilin no longer binds to actin (Meng et al., 2004). Phosphorylation of Cofilin

Discussion

The work presented here contributes several important points to our understanding of estrogen effects on spine synapse plasticity. First, these data demonstrate that estrogen controls the phospho-modulation of ADF/Cofilin via LIMK1, thus supporting that estrogen can act on the actin filament network to remodel neuron morphology. Second, the polymerization of actin filaments associated with Cofilin phosphorylation may explain how estrogen concomitantly drives the actin-based projections of

Materials

Rabbit polyclonal antibodies raised against phospho-Cofilin (p-Cofilin), total Cofilin (t-Cofilin), phospho-LIMK1/2 (p-LIMK), total LIMK, and β-actin were all purchased from Cell Signaling Technology (Beverly, MA). As stated by the manufacturer, this anti-pLIMK polyclonal antibody requires exogenous, recombinant protein for phospho-detection. The eGFP-chimeric wild-type (wt) LIMK-1 and eGFP-chimeric dnLIMK-1 plasmid constructs were kindly provided by Kensaku Mizuno, PhD (Tohoku University,

Acknowledgments

This study was supported in part by the NIH NS 07080 and P01 AG 16765 (BSMc and KTA). Additional support for this work comes from the NIH MSTP grant GM07739, The Surdna Foundation Medical Scientist Fellowship, and the Barbara and Stephen Friedman Fellowship Endowment (GSY).

References (71)

  • S.J. Lee et al.

    Estradiol affects spinophilin protein differently in gonadectomized males and females

    Neuroscience

    (2004)
  • B.S. McEwen et al.

    Hippocampal formation: shedding light on the influence of sex and stress on the brain

    Brain Res. Rev.

    (2007)
  • Y. Meng et al.

    Abnormal spine morphology and enhanced LTP in LIMK-1 knockout mice

    Neuron

    (2002)
  • Y. Meng et al.

    Regulation of ADF/Cofilin phosphorylation and synaptic function by LIM-kinase

    Neuropharmacol.

    (2004)
  • M. Nishio et al.

    Subcellular localization of estrogen receptor beta in mouse hippocampus

    Neurosci. Lett.

    (2004)
  • B. Racz et al.

    Spatial organization of Cofilin in dendritic spines

    Neuroscience

    (2006)
  • P.D. Sarmiere et al.

    Head, neck, and spines: a role for LIMK-1 in the hippocampus

    Neuron

    (2002)
  • M. Segal et al.

    Estradiol induces formation of dendritic spines in hippocampal neurons: functional correlates

    Horm. Behav.

    (2001)
  • Y. Sekino et al.

    Role of actin cytoskeleton in dendritic spine morphogenesis

    Neurochem. Int.

    (2007)
  • T. Shirao et al.

    Clustering and anchoring mechanisms of molecular constituents of postsynaptic scaffolds in dendritic spines

    Neurosci. Res.

    (2001)
  • J.L. Spencer et al.

    Uncovering the mechanisms of estrogen effects on hippocampal function

    Front. Neuroendocrinol.

    (2008)
  • J.L. Spencer et al.

    Estrous cycle regulates activation of hippocampal Akt, LIM kinase, and neurotrophin receptors in C57BL/6 mice

    Neuroscience

    (2008)
  • C.A. Stanyon et al.

    LIM-kinase1

    Int. J. Biochem. Cell Biol.

    (1999)
  • T. Sumi et al.

    Specific activation of LIM kinase 2 via phosphorylation of threonine 505 by ROCK, a Rho-dependent protein kinase

    J. Biol. Chem.

    (2001)
  • T. Tojima et al.

    Signal transduction cascades underlying de novo protein synthesis required for neuronal morphogenesis in differentiating neurons

    Prog. Neurobiol.

    (2004)
  • T. Tojima et al.

    Dual regulation of LIM kinase 1 expression by cyclic AMP and calcium determines Cofilin phosphorylation states during neuritogenesis in NG108-15 cells

    Brain Res.

    (2003)
  • M. Yildirim et al.

    Estrogen and aging affect synaptic distribution of phosphorylated LIM kinase (pLIMK) in CA1 region of female rat hippocampus

    Neuroscience

    (2008)
  • M. Yildirim et al.

    Effects of estrogen and aging on the synaptic distribution of phosphorylated Akt-immunoreactivity in the CA1 region of the female rat hippocampus

    Brain Res.

    (2011)
  • M.M. Adams et al.

    Estrogen and the aging hippocampal synapse

    Cereb. Cortex

    (2003)
  • K.T. Akama et al.

    Estrogen stimulates postsynaptic density-95 rapid protein synthesis via the Akt/protein kinase B pathway

    J. Neurosci.

    (2003)
  • D.W. Allison et al.

    Role of actin in anchoring postsynaptic receptors in cultured hippocampal neurons: differential attachment of NMDA versus AMPA receptors

    J. Neurosci.

    (1998)
  • T. Amano et al.

    LIM-kinase 2 induces formation of stress fibres, focal adhesions and membrane blebs, dependent on its activation by Rho-associated kinase-catalysed phosphorylation at threonine-505

    Biochem. J.

    (2001)
  • S. Arber et al.

    Regulation of actin dynamics through phosphorylation of Cofilin by LIM-kinase

    Nature

    (1998)
  • J.R. Bamburg

    Proteins of the ADF/Cofilin family: essential regulators of actin dynamics

    Annu. Rev. Cell Dev. Biol.

    (1999)
  • J.R. Bamburg et al.

    ADF/Cofilin-actin rods in neurodegenerative diseases

    Curr. Alzheimer Res.

    (2010)
  • Cited by (46)

    • Synaptic effects of estrogen

      2020, Vitamins and Hormones
      Citation Excerpt :

      Estrogens have also been shown to increase activation of the LIM kinase (LIMK) pathways in the female hippocampus through a mechanism involving estrogen-mediated phosphorylation of Cofilin, an actin depolymerization factor. This results in structural reorganization of the actin cytoskeletal network, resulting in filopodial extension and formation of new spines (Yuen, McEwen, & Akama, 2011). Notch signaling has been implicated in the inhibition of dendrite extension and synaptic connectivity (Berezovska et al., 1999; Salama-Cohen, Arevalo, Grantyn, & Rodriguez-Tebar, 2006; Sestan, Artavanis-Tsakonas, & Rakic, 1999).

    • Estradiol induces synaptic rearrangements

      2020, Vitamins and Hormones
      Citation Excerpt :

      Both RhoA kinase and PAK can stimulate LIMK1 activity (Dan, Kelly, Bernard, & Minden, 2001; Edwards, Sanders, Bokoch, & Gill, 1999; Maekawa et al., 1999), which inactivates cofilin. Inactivation of cofilin by LIMK-mediated phosphorylation at Ser 3 is a necessary step for the activation of actin polymerization (Yuen et al., 2011). Blocking of RhoA–RhoA kinase–LIM kinase (LIMK)–cofilin pathway eliminates E2 effects on spines and long-term potentiation (Kramar et al., 2009; Spencer, Waters, Milner, & McEwen, 2008; Yildirim et al., 2008; Yuen et al., 2011).

    View all citing articles on Scopus
    View full text