Elsevier

Cellular Signalling

Volume 63, November 2019, 109357
Cellular Signalling

Review
mAKAPβ signalosomes – A nodal regulator of gene transcription associated with pathological cardiac remodeling

https://doi.org/10.1016/j.cellsig.2019.109357Get rights and content

Highlights

  • Muscle A-kinase anchoring protein β (mAKAPβ) is a scaffolding protein localized to the nuclear envelope in striated muscle

  • Expression of mAKAPβ contributes to the induction of pathological hypertrophy

  • mAKAPβ orchestrates the regulation of gene transcription required for induction of cardiac hypertrophy

  • mAKAPβ regulates the activity of class IIa HDAC enzymes as well as the transcription factors NFAT, MEF2, and HIF-1α

Abstract

Striated myocytes compose about half of the cells of the heart, while contributing the majority of the heart's mass and volume. In response to increased demands for pumping power, including in diseases of pressure and volume overload, the contractile myocytes undergo non-mitotic growth, resulting in increased heart mass, i.e. cardiac hypertrophy. Myocyte hypertrophy is induced by a change in the gene expression program driven by the altered activity of transcription factors and co-repressor and co-activator chromatin-associated proteins. These gene regulatory proteins are subject to diverse post-translational modifications and serve as nuclear effectors for intracellular signal transduction pathways, including those controlled by cyclic nucleotides and calcium ion. Scaffold proteins contribute to the underlying architecture of intracellular signaling networks by targeting signaling enzymes to discrete intracellular compartments, providing specificity to the regulation of downstream effectors, including those regulating gene expression. Muscle A-kinase anchoring protein β (mAKAPβ) is a well-characterized scaffold protein that contributes to the regulation of pathological cardiac hypertrophy. In this review, we discuss the mechanisms how this prototypical scaffold protein organizes signalosomes responsible for the regulation of class IIa histone deacetylases and cardiac transcription factors such as NFAT, MEF2, and HIF-1α, as well as how this signalosome represents a novel therapeutic target for the prevention or treatment of heart failure.

Introduction

An increase in the mass of the heart known as “cardiac hypertrophy” is the primary response of that organ to stress in disease and a major risk factor for the development of heart failure, a leading cause of death and a problem of increasing public health significance worldwide [1,2]. According to the latest statistics from the American Heart Association, an estimated 6.2 million Americans have heart failure [3]. This number is estimated to increase 46% by 2030. Furthermore, the 5-year mortality rate of patients diagnosed with heart failure is ~50%, demonstrating the need for better therapeutics to treat this syndrome. The main contributor to the increased heart size of the hypertrophic heart is hypertrophy of the cardiac myocytes, an increase in the volume of the contractile cells in the absence of cellular proliferation. Fundamental to the development of hypertrophy is a dramatic altering of the myocyte gene expression program, that besides the change in morphology also results in disease in changes in cellular metabolism, contractility, and survival, and the release of paracrine factors that promote myocardial interstitial fibrosis [4]. Research over the last 20 years has revealed that select transcription factors, including nuclear factor of activated T-cells (NFAT) and myocyte enhancer factor 2 (MEF2) family members, nucleate the chromatin activating and repressive transcriptional complexes that direct pathological cardiac gene expression [4]. In addition, changes in the activity of histone deactylases (HDACs) and other chromatin modifying enzymes are integral to the induction of hypertrophy [5]. As these gene regulatory proteins are tightly regulated by intracellular signal transduction pathways, the elucidation of the upstream signals that control transcription factor and epigenetic modifier activity remains an important area of heart failure research. Extensive research has shown the relevance of mitogen-activated protein kinase (MAPK), cyclic nucleotide, calcium, hypoxia and phosphoinositide-dependent pathways to the regulation of gene regulatory protein post-translational modification in hypertrophy [6]. However, the mechanisms of how these pathways selectively control relevant gene regulatory proteins in disease remains unclear, especially given the separate role that cyclic nucleotides and calcium play in the regulation of excitation-contraction coupling.

One mechanism that has evolved to confer specificity to signaling networks is compartmentation by scaffolding proteins [7]. These typically non-enzymatic proteins function to co-localize signaling enzymes into discrete complexes with both their upstream activators and their downstream effector substrates, providing enhanced substrate specificity while increasing the kinetics of signaling events. In addition, as scaffold proteins are often multivalent and bind to enzymes from multiple signaling pathways, “signalosomes” organized by scaffold proteins serve to integrate multiple upstream signals and facilitate crosstalk between different relevant signaling pathways, thereby providing a combinatorial regulation of downstream signaling events. As discussed herein, research concerning the scaffold protein muscle A-kinase anchoring protein β (mAKAPβ) has revealed that mAKAPβ signalosomes are central to the orchestration of gene regulatory proteins controlling pathological cardiac remodeling.

A-Kinase anchoring proteins (AKAPs) are scaffold proteins that bind the cAMP-dependent Protein Kinase A (PKA) [8]. Directing PKA to discrete cellular compartments via localization domains unique to each AKAP and binding multiple other signaling enzymes such as phosphatases, phosphodiesterases, and other kinases, AKAPs serve important roles in the facilitation of cross-talk between different signaling pathways, including G-protein coupled receptor signaling in the heart [9]. The first demonstration of the importance of AKAPs for cardiac physiology utilized a peptide to globally disrupt AKAP/PKA binding [10]. Incubation of cardiac myocytes with a PKA anchoring disruptor peptide showed that AKAPs play a role in the β-adrenergic stimulation of calcium influx and contraction. Since these original findings, AKAP have been shown to regulate many cellular events such as potassium channel currents, sarcoplasmic calcium cycling, and G-protein coupled signaling [[11], [12], [13]]. Additionally, several AKAPs are involved in the induction of cardiac disease [9]. This review will focus on the function of mAKAPβ signalosomes in the induction of pathological cardiac hypertrophy through the orchestrated regulation of myocyte gene expression.

Section snippets

mAKAP – a perinuclear scaffold protein that regulates cardiac myocyte hypertrophy

The mAKAP (AKAP6) gene is alternatively-spliced and expressed as the 260 kDa α-isoform in neurons and the 230 kDa β-isoform in striated myocytes, such that the mAKAPβ protein is identical to mAKAPα aa residues 245–2314 due to initiation of translation at an internal residue (Fig. 1A) [14,15]. Although initially misconstrued to be localized to the sarcoplasmic reticulum [16,17], in terminally differentiated striated myocytes, mAKAPβ is constitutively localized in myocytes to the nuclear envelope

mAKAP signalosomes and regulation of class IIa HDACs

When present in the nucleus, class IIa HDACs (HDACs 4, 5, 7, and 9), which have weak intrinsic HDAC activity, organize co-repressor complexes that suppress the activity of site-specific transcription factors [34,35]. Nuclear export of class IIa HDACs permits the expression of genes activated by the MEF2 family of transcription factors key to the development of pathological cardiac remodeling, while in normal cardiac physiology class IIa HDAC-mediated repression maintains normal cardiac

mAKAPβ and NFAT

The NFAT family of transcription factors, in particular NFATc2 and NFATc3, are key regulators of cardiac hypertrophy (Fig. 5A) [65,66]. They are present in the cytoplasm of resting cells due to multiple sites of phosphorylation, but translocate to the nucleus following dephosphorylation by calcineurin [67]. The mAKAP complex has been shown to regulate the activity of multiple members of the NFAT family. NFATc3 co-precipitates with mAKAPβ isolated from rat cardiac extracts, although it may not

mAKAPβ REGULATION OF MEF2

The myocyte enhancer factor 2 (MEF2) family of transcription factors (MEF2A, MEF2B, MEF2C and MEF2D) are important for both heart development and induction of cardiac remodeling, as well as skeletal myoblast differentiation [[68], [69], [70]]. For example, conditional MEF2D gene deletion attenuated pathological cardiac remodeling in response to pressure overload and chronic catecholamine infusion [71]. MEF2D is an effector for mAKAPβ signalosomes, and MEF2D-mAKAPβ complexes can be isolated from

HIF-1-α and mAKAPβ

The central role for mAKAPβ in hypertrophic gene transcription has raised the question whether mAKAPβ can regulate additional transcription factors involved in other forms of heart disease. Biochemical studies have revealed that mAKAPβ binds the transcription factor hypoxia-inducible factor 1-alpha (HIF-1α) and related regulatory proteins in cardiac myocytes (Fig. 6) [78]. This basic helix-loop PAS domain containing protein is a critical regulator of the cellular response to hypoxia [79]. In

A role for mAKAPβ in intranuclear PKA regulation?

Most of the research concerning mAKAPβ has been premised upon the idea that regulation of mAKAPβ effector proteins occurs within a discrete perinuclear compartment. It is possible however, that mAKAPβ-associated PKA might have effects distal from the scaffold. Work performed in adult rat ventricular myocytes found that a nuclear pool of cAMP was activated by both β1- and β2-adrenergic receptors, while only the former receptor increased nucleoplasmic PKA activity and the expression of the gene

Perspective

It is well-established in the basic cardiovascular sciences literature that induction of pathological cardiac remodeling requires an altered myocyte gene expression program. How chromatin-associated factors such as MEF2, NFAT, HIF-1α, and class IIa HDACs are individually regulated has been well-studied, but much remains to be understood regarding their coordinated regulation by upstream second messenger systems. The discovery of mAKAPβ signalosomes has provided a unique insight into the

Acknowledgments

This work was funded, in whole or in part, by National Institutes of Health Grants HL126825 and HL146111 (K.D.K. and M.S.K.) and HL126950 and EY026766 (M.S.K), California Tobacco-Related Disease Research Grants Program Office of the University of California, Grant Number No. 27IR-0045 (M.S.K.), and by an American Heart Association Predoctoral Grant 18PRE34030209 to MG. The opinions, findings, and conclusions herein are those of the authors and not necessarily represent those The Regents of the

Delcaration of Competing Interest

Dr. Kapiloff owns equity in Anchored RSK3 Inhibitors, LLC, and Cardiac RSK3 Inhibitors, LLC, companies interested in developing mAKAPβ-based therapies.

References (90)

  • C.L. Zhang

    Class II histone deacetylases act as signal-responsive repressors of cardiac hypertrophy

    Cell

    (2002)
  • G.K. Carnegie

    AKAP-Lbc mobilizes a cardiac hypertrophy signaling pathway

    Mol. Cell

    (2008)
  • C.W. Chang

    Acute beta-adrenergic activation triggers nuclear import of histone deacetylase 5 and delays G(q)-induced transcriptional activation

    J. Biol. Chem.

    (2013)
  • C.C. Sucharov

    beta-Adrenergic receptor stimulation and activation of protein kinase A protect against alpha1-adrenergic-mediated phosphorylation of protein kinase D and histone deacetylase 5

    J. Card. Fail.

    (2011)
  • K.L. Dodge-Kafka

    Bidirectional regulation of HDAC5 by mAKAPbeta signalosomes in cardiac myocytes

    J. Mol. Cell. Cardiol.

    (2018)
  • M.S. Kapiloff

    An adenylyl cyclase-mAKAPbeta signaling complex regulates cAMP levels in cardiac myocytes

    J. Biol. Chem.

    (2009)
  • C. Sette et al.

    Phosphorylation and activation of a cAMP-specific phosphodiesterase by the cAMP-dependent protein kinase. Involvement of serine 54 in the enzyme activation

    J. Biol. Chem.

    (1996)
  • K.L. Dodge-Kafka

    cAMP-stimulated protein phosphatase 2A activity associated with muscle a kinase-anchoring protein (mAKAP) signaling complexes inhibits the phosphorylation and activity of the cAMP-specific phosphodiesterase PDE4D3

    J. Biol. Chem.

    (2010)
  • A. Rababa’h

    Protein kinase a and phosphodiesterase-4D3 binding to coding polymorphisms of cardiac muscle anchoring protein (mAKAP)

    J. Mol. Biol.

    (2013)
  • L. Zhang

    Phospholipase Cepsilon hydrolyzes perinuclear phosphatidylinositol 4-phosphate to regulate cardiac hypertrophy

    Cell

    (2013)
  • J. Li

    The mAKAPbeta scaffold regulates cardiac myocyte hypertrophy via recruitment of activated calcineurin

    J. Mol. Cell. Cardiol.

    (2010)
  • M. Bourajjaj

    NFATc2 is a necessary mediator of calcineurin-dependent cardiac hypertrophy and heart failure

    J. Biol. Chem.

    (2008)
  • B.J. Wilkins et al.

    Calcium-calcineurin signaling in the regulation of cardiac hypertrophy

    Biochem. Biophys. Res. Commun.

    (2004)
  • J. Li

    Regulation of MEF2 transcriptional activity by calcineurin/mAKAP complexes

    Exp. Cell Res.

    (2013)
  • S. Gregoire

    Control of MEF2 transcriptional activity by coordinated phosphorylation and sumoylation

    J. Biol. Chem.

    (2006)
  • J. Li

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

    J. Biol. Chem.

    (2019 Feb 15)
  • N.T. Buu et al.

    Norepinephrine in neonatal rat ventricular myocytes: association with the cell nucleus and binding to nuclear alpha 1- and beta-adrenergic receptors

    J. Mol. Cell. Cardiol.

    (1993)
  • G. Iwami

    Regulation of adenylyl cyclase by protein kinase A

    J. Biol. Chem.

    (1995)
  • J.C. Madukwe

    G protein betagamma subunits directly interact with and activate phospholipase C

    J. Biol. Chem.

    (2018)
  • J. Li

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

    J. Biol. Chem.

    (2019)
  • J.S. Burchfield et al.

    Pathological ventricular remodeling: mechanisms: part 1 of 2

    Circ.

    (2013)
  • E.J. Benjamin

    Heart Disease and Stroke Statistics-2018 Update: A Report From the American Heart Association

    Circ.

    (2018)
  • E.J. Benjamin

    Heart disease and stroke Statistics-2019 update: a report from the American Heart Association

    Circ.

    (2019)
  • J.H. van Berlo et al.

    Signaling effectors underlying pathologic growth and remodeling of the heart

    J. Clin. Invest.

    (2013)
  • T.A. McKinsey et al.

    Epigenomic regulation of heart failure: integrating histone marks, long noncoding RNAs, and chromatin architecture

    F1000Res

    (2018)
  • J. Heineke et al.

    Regulation of cardiac hypertrophy by intracellular signalling pathways

    Nat. Rev. Mol. Cell Biol.

    (2006)
  • J.D. Scott et al.

    Cell signaling in space and time: where proteins come together and when they’re apart

    Sci.

    (2009)
  • J.D. Scott et al.

    Creating order from chaos: cellular regulation by kinase anchoring

    Annu. Rev. Pharmacol. Toxicol.

    (2013)
  • D. Diviani

    A-kinase anchoring proteins: scaffolding proteins in the heart

    Am. J. Physiol. Heart Circ. Physiol.

    (2011)
  • M.A. Fink

    AKAP-mediated targeting of protein kinase a regulates contractility in cardiac myocytes

    Circ. Res.

    (2001)
  • S.O. Marx

    Requirement of a macromolecular signaling complex for beta adrenergic receptor modulation of the KCNQ1-KCNE1 potassium channel

    Sci.

    (2002)
  • B. Lygren

    AKAP complex regulates Ca2+ re-uptake into heart sarcoplasmic reticulum

    EMBO Rep.

    (2007)
  • M.S. Kapiloff

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

    J. Cell Sci.

    (1999)
  • J.J. Michel

    Spatial restriction of PDK1 activation cascades by anchoring to mAKAPalpha

    Mol. Cell

    (2005)
  • M.D. Kritzer

    The scaffold protein muscle A-kinase anchoring protein beta orchestrates cardiac myocyte hypertrophic signaling required for the development of heart failure

    Circ. Heart Fail.

    (2014)
  • Cited by (20)

    • A cellular atlas of calcineurin signaling

      2023, Biochimica et Biophysica Acta - Molecular Cell Research
      Citation Excerpt :

      AKAP6, also known as mAKAPβ, is expressed primarily in striated muscle including cardiomyocytes, and associates with the nuclear envelope via a cluster of spectrin-like repeats [154]. AKAP6 directly scaffolds multiple proteins including RyR, calcineurin (the CNAβ isoform), PKA and MEF2, and is required for CN-dependent regulation of NFAT and MEF2 that promotes cardiac hypertrophy in response to β-adrenergic signaling [154]. Thus, AKAP6 is proposed to organize a ‘signalosome’ at the nuclear envelope that locally organizes and integrates CN activity with other signaling pathways during cardiac hypertrophy.

    • A perinuclear calcium compartment regulates cardiac myocyte hypertrophy

      2022, Journal of Molecular and Cellular Cardiology
      Citation Excerpt :

      Together, these results suggest that perinuclear cAMP-PKA signaling is required for activation of the CaN-NFAT pathway that regulates cardiac myocyte hypertrophy. Biochemical and physiological studies have implicated the perinuclear mAKAPβ signalosome in the regulation of gene expression responsible for cardiac myocyte hypertrophy [9]. Live cell imaging experiments using novel tools for compartment-specific signaling modulation now provide evidence that mAKAPβ organizes an independent Ca2+ signaling compartment that regulates CaN and NFAT transcription factor required for hypertrophy (Fig. 8).

    • Calcineurin in the heart: New horizons for an old friend

      2021, Cellular Signalling
      Citation Excerpt :

      Mice with a cardiomyocyte-specific deletion of AKAP6 are resistant to both pressure overload and agonist induced heart failure [234]. Extensive details of the many signaling processes mediated through AKAP6 and their relevance to cardiac remodeling can be found in an excellent recent review [237]. AKAP1 (AKAP121/AKAP84), located at the outer mitochondrial membrane (OMM), acts as a coordinating hub for mitochondrial dynamics driven by changes in phosphorylation of the Dynamin Related Protein 1 (DRP1/DNM1L).

    • From classical signaling pathways to the nucleus

      2021, Epigenetics in Cardiovascular Disease
    View all citing articles on Scopus
    View full text