Original article
The mAKAPβ scaffold regulates cardiac myocyte hypertrophy via recruitment of activated calcineurin

https://doi.org/10.1016/j.yjmcc.2009.10.023Get rights and content

Abstract

mAKAPβ is the scaffold for a multimolecular signaling complex in cardiac myocytes that is required for the induction of neonatal myocyte hypertrophy. We now show that the pro-hypertrophic phosphatase calcineurin binds directly to a single site on mAKAPβ that does not conform to any of the previously reported consensus binding sites. Calcineurin–mAKAPβ complex formation is increased in the presence of Ca2+/calmodulin and in norepinephrine-stimulated primary cardiac myocytes. This binding is of functional significance because myocytes exhibit diminished norepinephrine-stimulated hypertrophy when expressing a mAKAPβ mutant incapable of binding calcineurin. In addition to calcineurin, the transcription factor NFATc3 also associates with the mAKAPβ scaffold in myocytes. Calcineurin bound to mAKAPβ can dephosphorylate NFATc3 in myocytes, and expression of mAKAPβ is required for NFAT transcriptional activity. Taken together, our results reveal the importance of regulated calcineurin binding to mAKAPβ for the induction of cardiac myocyte hypertrophy. Furthermore, these data illustrate how scaffold proteins organizing localized signaling complexes provide the molecular architecture for signal transduction networks regulating key cellular processes.

Introduction

Cardiac myocyte hypertrophy is the major intrinsic mechanism by which the heart may counterbalance chronically elevated demands for pumping power. Myocyte hypertrophy is controlled by a network of intracellular signaling pathways that are activated by G-protein coupled, growth factor and cytokine receptors and by mechanical and oxidative stress [1]. These signals are transduced by MAPK, cyclic nucleotide, Ca2+ and phosphoinositide-dependent pathways. Although much progress has been made over the last 20 years to define this network, it is still unclear how the various constituent pathways act in concert to regulate the overall cellular phenotype [2]. Moreover, while individual signaling pathways may regulate specific cellular functions, the molecules that comprise these signaling pathways often serve multiple functions in the same cells. Therefore, an important question in the field of signal transduction has been how pleiotropic signaling molecules such as protein kinases and phosphatases can specifically regulate individual downstream effectors in response to different upstream stimuli. One mechanism by which specificity in signal transduction is conferred is the formation of multimolecular signaling complexes by scaffold proteins of different combinations of common signaling enzymes [3].

While signaling enzymes may be broadly distributed within the cell, scaffold proteins, such as A-kinase anchoring proteins (AKAPs), recruit small pools of these enzymes to discrete multimolecular complexes that are sequestered in distinct intracellular compartments and that serve different cellular functions [4]. mAKAP (muscle AKAP) was initially identified in a screen for protein kinase A (PKA) binding proteins. mAKAPα and mAKAPβ are the two known isoforms encoded by the single mAKAP (AKAP6) gene and are expressed in neurons and striated myocytes, respectively [5]. As a consequence of alternative mRNA splicing, mAKAPβ is identical to residues 245–2314 (the C-terminus) of mAKAPα. In adult and neonatal cardiac myocytes, mAKAPβ is primarily localized to the outer nuclear membrane through its association with nesprin-1α [6], [7]. In addition to PKA, proteins that have been shown to associate with the mAKAPβ scaffold in myocytes include adenylyl cyclase type 5 [8], the cAMP-specific phosphodiesterase PDE4D3 [9], the cAMP-activated guanine nucleotide exchange factor Epac1 [10], ERK5 and MEK5 mitogen-activated protein kinases (MAPK) [10], the Ca2+/calmodulin-dependent protein phosphatase calcineurin Aβ (CaN, PP2B) [11], protein phosphatase 2A [12], hypoxia-inducible factor 1α (HIF1α) and ubiquitin E3-ligases involved in HIF1α regulation [13], myopodin [14], the ryanodine receptor Ca2+-release channel (RyR2) [12], [15] and the sodium/calcium exchanger NCX1 [16]. Due to the association of these various enzymes and ion channels with mAKAPβ in the cardiac myocyte, we have proposed that mAKAPβ complexes are important for the regulation of pathologic myocyte remodeling in response to upstream cAMP, calcium, and MAPK signals and hypoxic stress [13], [17]. In support of this hypothesis, mAKAPβ expression in myocytes is required for the full induction of neonatal myocyte hypertrophy in vitro by adrenergic and cytokine agonists [10], [11].

CaN is a pleiotropic Ca2+/calmodulin-dependent serine/threonine phosphatase composed of a catalytic A-subunit and a regulatory B-subunit [18]. There are three mammalian A-subunits, of which Aα and Aβ are expressed ubiquitously and Aγ is restricted to testes. Aα and Aβ have been studied by genetic deletion and are not functionally redundant. For example, only the CaNAβ isoform is important for the induction of pathologic cardiac hypertrophy and the survival of myocytes after ischemia [19], [20]. Important calcineurin substrates in vivo include four of the five members of the nuclear factor of activated T-cell transcription factor family (NFATc 1–4). In addition to forming heterodimers with other transcription factors, NFATc can bind directly to CaN through conserved PxIxIT and LxVP motifs [21]. CaN binding facilitates dephosphorylation of the N-terminal NFATc regulatory domain, inducing NFATc nuclear translocation from the cytoplasm. Accordingly, NFATc isoforms serve important roles in cardiac development and myocyte hypertrophy [22].

Previously, we showed that CaNAβ is associated with mAKAPβ in cardiac myocytes [11]. However, it remains unclear how scaffolding by this relatively low abundant protein contributes to CaN signaling. In this study, we characterize the direct binding of CaNAβ to mAKAPβ. Moreover, we provide evidence that recruitment of CaNAβ and NFATc3 to mAKAPβ complexes is important for the transduction of hypertrophic signaling.

Section snippets

Antibodies and antiserum

Commercially available antibodies were as follows: rabbit and mouse anti-Flag (Sigma), anti-S tag (Novagen), anti-His tag (Santa Cruz), mouse anti-HA tag (Sigma), mouse anti-myc tag (monoclonal 4A6, Millipore), rabbit anti-CaNAβ (Santa Cruz), mouse anti-NFATc1 (BD Biosciences), rabbit anti-NFATc3 (Santa Cruz), rabbit anti-CaNAβ (Santa Cruz), mouse anti-α-actinin (monoclonal EA-53, Sigma), rabbit anti-rat atrial natriuretic factor (ANF; US Biological), horseradish peroxidase (HRP)-conjugated

Binding of mAKAPβ and CaN in myocytes

As we have shown previously [11], endogenous CaNAβ and mAKAPβ associated in adult rat heart extracts can be co-immunoprecipitated with a mAKAPβ-specific antibody (Fig. 1B). In order to test whether mAKAPβ can also bind CaNAα, HEK293 cells were transfected with expression vectors for mAKAPβ and either CaNAα or CaNAβ HA-tagged isoforms (Fig. 1C). mAKAPβ was efficiently co-immunoprecipitated when either HA-tagged CaN isoform was present (lanes 2 and 3). In addition, mAKAPβ was

Discussion

In this paper we present evidence that CaNAβ forms inducible complexes with the scaffold protein mAKAPβ that are important for the transduction of hypertrophic signaling. In contrast to CaN scaffolds that have been previously described such as calsarcin, AKAP79/150 and AKAP121 which inhibit CaN activity and may compete for NFATc binding [25], [26], [27], CaNAβ bound to a mAKAPβ fusion protein actively dephosphorylated phospho-NFATc3 substrate (Fig. 5C). Moreover, CaNAβ–mAKAPβ binding was

Acknowledgments

This work is supported by the National Heart, Lung, and Blood Institute grant RO1 HL075398 to M.S.K. and HL082705 to K.D.K.

References (36)

  • T. Pawson et al.

    Assembly of cell regulatory systems through protein interaction domains

    Science

    (2003 Apr 18)
  • G.K. Carnegie et al.

    A-kinase anchoring proteins: from protein complexes to physiology and disease

    IUBMB Life

    (2009 Apr)
  • J.J. Michel et al.

    Spatial restriction of PDK1 activation cascades by anchoring to mAKAPalpha

    Mol. Cell.

    (2005 Dec 9)
  • M.S. Kapiloff et al.

    mAKAP: an A-kinase anchoring protein targeted to the nuclear membrane of differentiated myocytes

    J. Cell. Sci.

    (1999)
  • K.L. Dodge et al.

    mAKAP assembles a protein kinase A/PDE4 phosphodiesterase cAMP signaling module

    EMBO J.

    (2001 Apr 17)
  • K.L. Dodge-Kafka et al.

    The protein kinase A anchoring protein mAKAP coordinates two integrated cAMP effector pathways

    Nature

    (2005 Sep 22)
  • G.C. Pare et al.

    The mAKAP complex participates in the induction of cardiac myocyte hypertrophy by adrenergic receptor signaling

    J. Cell. Sci.

    (2005 Dec 1)
  • M.S. Kapiloff et al.

    mAKAP and the ryanodine receptor are part of a multi-component signaling complex on the cardiomyocyte nuclear envelope

    J. Cell. Sci.

    (2001 Sep)
  • Cited by (58)

    • A perinuclear calcium compartment regulates cardiac myocyte hypertrophy

      2022, Journal of Molecular and Cellular Cardiology
      Citation Excerpt :

      Iso induced a CaNAR2-nesprin transient in neonatal myocytes that was blocked by the CaN inhibitor cyclosporin A (CsA, Fig. 1G). The increase in perinuclear CaN activity was relatively slow in onset consistent with the relatively slow recruitment of CaN to the mAKAPβ scaffold in stimulated myocytes [14]. Like the perinuclear Ca2+ transient, CaNAR2-nesprin signal was dependent upon mAKAPβ expression in neonatal myocytes (Fig. 1H).

    • Mapping genetic changes in the cAMP-signaling cascade in human atria

      2021, Journal of Molecular and Cellular Cardiology
    • mAKAPβ signalosomes – A nodal regulator of gene transcription associated with pathological cardiac remodeling

      2019, Cellular Signalling
      Citation Excerpt :

      mAKAPβ-bound PKA can phosphorylate a small pool of perinuclear type II ryanodine receptors (RyR) also associated with mAKAPβ and nesprin-1α (Fig. 5) [17,19,22]. Calcium released from perinuclear stores through this channel is likely responsible for local activation of the Ca2+/calmodulin phosphatase calcineurin (PP2B, CaN) that binds a discrete domain within mAKAPβ (aa 1286–1345 [64]) and that can in turn induce NFAT and MEF2-dependent gene expression associated with hypertrophy [64], as discussed in the following sections. The NFAT family of transcription factors, in particular NFATc2 and NFATc3, are key regulators of cardiac hypertrophy (Fig. 5A) [65,66].

    • Muscle A-kinase–anchoring protein- bound calcineurin toggles active and repressive transcriptional complexes of myocyte enhancer factor 2D

      2019, Journal of Biological Chemistry
      Citation Excerpt :

      Unlike at other scaffolds where CaN is bound in an inactive conformation ready for local action (22), we found that catalytically active CaN is recruited to mAKAPβ upon myocyte stimulation, whether in cardiac myocytes with norepinephrine (13) or as shown now in C2C12 cells with DM (Fig. 1). Binding of CaN to mAKAPβ was required for NFATc3 activation in cardiac myocytes (13) and MEF2D dephosphorylation at Ser-444 (Fig. 2). It has been reported that CaN translocates into the nucleus with NFAT transcription factors when activated (25).

    View all citing articles on Scopus
    1

    These authors contributed equally to this manuscript.

    View full text