molecular pharmacology fast forward. published on december ...€¦ · 2/12/2013  · underlie...

52
MOL #90365 1 MAPK-activated protein kinase 2 (MK2) in neuroinflammation, Hsp27 phosphorylation and cell cycle: Role and Targeting Fadi Maged Shokry Gurgis, William Ziaziaris, Lenka Munoz Department of Pharmacology, School of Medical Sciences, University of Sydney, NSW 2006, Australia (FMSG, WZ, LM) Molecular Pharmacology Fast Forward. Published on December 2, 2013 as doi:10.1124/mol.113.090365 Copyright 2013 by the American Society for Pharmacology and Experimental Therapeutics. This article has not been copyedited and formatted. The final version may differ from this version. Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365 at ASPET Journals on July 5, 2021 molpharm.aspetjournals.org Downloaded from This article has not been copyedited and formatted. The final version may differ from this version. Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365 at ASPET Journals on July 5, 2021 molpharm.aspetjournals.org Downloaded from

Upload: others

Post on 15-Feb-2021

0 views

Category:

Documents


0 download

TRANSCRIPT

  • MOL #90365

    1

    MAPK-activated protein kinase 2 (MK2) in neuroinflammation, Hsp27 phosphorylation

    and cell cycle: Role and Targeting

    Fadi Maged Shokry Gurgis, William Ziaziaris, Lenka Munoz

    Department of Pharmacology, School of Medical Sciences, University of Sydney, NSW

    2006, Australia (FMSG, WZ, LM)

    Molecular Pharmacology Fast Forward. Published on December 2, 2013 as doi:10.1124/mol.113.090365

    Copyright 2013 by the American Society for Pharmacology and Experimental Therapeutics.

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    This article has not been copyedited and formatted. The final version may differ from this version.

    Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365 at A

    SPET

    Journals on July 5, 2021m

    olpharm.aspetjournals.org

    Dow

    nloaded from

    http://molpharm.aspetjournals.org/http://molpharm.aspetjournals.org/

  • MOL #90365

    2

    Running title: MK2, neuroinflammation and cell cycle

    Author for correspondence: Dr Lenka Munoz, Department of Pharmacology, School of Medical Sciences, University of Sydney, NSW 2006 Australia; e-mail: [email protected]

    Pages: 49 Abstract: 239 Figures: 3 Introduction: 324 (incl. referencing) References: 145 Discussion: 6,459 (incl. referencing)

    Abbreviations: AATF, apoptosis-antagonizing transcription factor; ARE, AU-rich element;

    ATM ataxia telangiectasia mutated; ATR, ataxia telangiectasia and Rad3 related; BE,

    biochemical efficiency; CAMK, calcium/calmodulin-dependent kinase; CDC25, cell

    division cycle phosphatase; CDK, cyclin-dependent kinase; COX2, cyclooxygenase 2; EMT,

    epithelial-to-mesenchymal transition; ERK, extracellular signal-regulated kinase; hnRNP,

    heterogeneous nuclear ribonucleoprotein; HNSCC, head and neck squamous cancer cells;

    HuR, human antigen R; Hsp, heat shock protein; Chk1/2, checkpoint kinase 1/2; IL,

    interleukin; IRF3, interferon regulatory factor 3; JNK, c-Jun N-terminal kinase; LIMK1,

    LIM-kinase 1; MAPK, mitogen-activated protein kinase; MMP, matrix metalloproteinases ;

    MK2/3, MAPK-activated protein kinase 2/3; MKK3, MAPK kinase 3; MPTP, 1-methyl-4-

    phenyl-1,2,3,6-tetrahydropyridine; MRLC3, myosin-regulatory light chain 3; NES, nuclear

    export sequence; NFκB, nuclear factor κB; NLS, nuclear localization signal; PABP1, polyA-

    binding protein 1; PARN, poly(A)specific ribonuclease; PKD, protein kinase D; Plk1, polo-

    like kinase 1; PMA, phorbol-12-myristate-13-acetate; PTEN, phosphatase and tensin

    homolog; RBP, RNA binding protein; SCI, spinal cord injury; SPARC, secreted protein

    acidic and rich in cysteine; STAT3, signal transducer and activator of transcription 3; TAK1,

    transforming growth factor β-activated kinase 1; TAO, thousand-and-one amino acid; TGFβ,

    transforming growth factor β; TMZ, temozolomide; TNFα, tumour necrosis factor α; TTP,

    tristetraproline; VEGF, vascular endothelial growth factor.

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    3

    Abstract

    Mitogen-activated protein kinase (MAPK)-activated protein kinase 2 (MAPKAPK-2 or

    MK2) is a downstream substrate of the p38 MAPK responsible for the signaling events

    influencing inflammation, cell division and differentiation, apoptosis as well as cell motility

    in response to a wide range of extracellular stimuli. Because of the failure of p38 MAPK

    inhibitors in clinical trials, MK2 unveiled as a potential target to regulate inflammatory

    cytokines mRNA stability and translation. Recent work suggests that this mechanism may

    underlie pathophysiology of brain disorders associated with inflammation. In addition, MK2

    is a prominent kinase that phosphorylates heat shock protein 27, an intensively investigated

    biomarker of cancer progression. This phosphorylation decreases its chaperone properties,

    making MK2 an endogenous inhibitor of Hsp27. MK2 is also one of the major players in the

    signal transduction pathways activated in response to DNA damage. Experimental evidence

    highlights the role of MK2 in G2/M and the mitotic spindle checkpoints, two mechanisms by

    which MK2 contributes to the maintenance of genomic stability. Thus, MK2 is considered a

    good molecular target to increase, in combination with chemotherapeutic agents, the

    sensitivity of treatment especially in p53-mutated tumors. This review looks at the functions

    of MK2 in inflammation, Hsp27 regulation and cell cycle checkpoint control with focus on

    brain pathologies. Analysis of MK2 signaling in various disease models and summary of the

    data on MK2 inhibitors suggest novel indications for MK2 inhibitors in addition to their

    mainstream use against peripheral inflammatory disorders.

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    4

    Introduction

    Mitogen-activated protein kinase (MAPK)-activated protein kinase 2 (MAPKAP-2 or MK2)

    is a downstream substrate of the p38 MAPK known to transduce a range of extracellular

    signals resulting in inflammatory response, cell division and differentiation, apoptosis as well

    as cell motility. Because of its central role in inflammation, p38 MAPK has been extensively

    investigated over the past decades. However, tremendous effort invested into the

    development of selective and efficacious p38 MAPK inhibitors has not delivered the much-

    needed small-molecule anti-inflammatory drug. As p38 MAPK regulates activity of more

    than 60 substrates (Trempolec et al., 2013), p38 MAPK inhibition is explicably accompanied

    with unwanted side effects and failure in clinical trials.

    MK2 is the main and essential down-stream target of p38 MAPK regulating

    biosynthesis of tumor necrosis factor α (TNFα) and other cytokines (Kotlyarov et al., 1999).

    Thus, MK2 emerged as a potential anti-inflammatory target, which is convincingly

    documented by increasing number of MK2-inflammation related publications and reviews. In

    addition, MK2 governs not only peripheral inflammation, but also the inflammation of the

    brain (termed neuroinflammation, (Culbert et al., 2006; Ghasemlou et al., 2010; Thomas et

    al., 2008b). MK2 is the prominent kinase phosphorylating heat shock protein 27 (Hsp27,

    Stokoe et al., 1992) that has turned as a promising target for cancer treatment. Finally, MK2

    is activated following DNA damage (Manke et al., 2005; Reinhardt et al., 2007), resulting in

    cell cycle arrest so that cells have the capacity to repair their DNA and continue to

    proliferate. Because this mechanism underlies resistance to chemotherapy, MK2 inhibitors

    could as serve as anti-cancer agents that improve efficacy of chemotherapy. As these aspects

    of MK2 biology have not been recently summarized, we present here a review on the role of

    MK2 in neuroinflammation-associated brain disorders. We will then discuss studies outlining

    the biological role of MK2-dependent regulation of Hsp27 activity and cell cycle control.

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    5

    Finally, we will conclude with therapeutic potential of MK2 inhibitors, and where possible

    focus on brain pathologies.

    Structure and function of MK2

    MK2 was discovered as an ERK1/2-activated protein kinase that had the capacity to

    phosphorylate and thus inactivate Hsp27 and its murine homologue Hsp25 (Stokoe et al.,

    1992). In following years, p38 MAPK was found to phosphorylate MK2 in response to stress

    stimuli. Further studies confirmed that MK2 activation depends on p38α/β MAPK-regulated

    phosphorylation of Thr222 in the activation loop, Ser272 in the catalytic domain and Thr334

    (Figure 1A) located in the hinge region between the catalytic domain and C-terminal

    regulatory domain (Ben-Levy et al., 1995).

    MK2 displays high homology (75% amino acid identity) to MK3 and their kinase

    domains are similar to other members of the calcium- and calmodulin-regulated kinases

    (CAMK) superfamily. MK2 expresses an N-terminal proline-rich region, which is

    responsible for interactions with Src homology 3 domains in vitro (Plath et al., 1994). The C-

    terminus contains a functional bipartite nuclear localisation signal (NLS) sequence that

    maintains the location of MK2 predominantly in the nuclei of resting cells (Figure 1).

    Conversely, the nuclear export sequence (NES) is located in the N-terminal to the NLS

    domain and triggers nuclear export following MK2 activation (Figure 1) (Ben-Levy et al.,

    1998; Engel et al., 1998).

    The crystal structure of MK2 revealed that Thr334 phosphorylation serves as a switch

    for MK2 nuclear import and export. Indeed, phosphomimetic mutation of Thr334 enhanced

    cytoplasmic localization of MK2, suggesting that MK2 contain a constitutively active NLS

    and a phosphorylation-regulated NES. In resting cells, p38 MAPK and MK2 form a complex

    in the nucleus (Figure 1B) ((Ben-Levy et al., 1998). Cellular stress causes the

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    6

    phosphorylation of p38 MAPK by upstream kinases such as MKK3. The activated p38

    MAPK then phosphorylates MK2 at Thr222, Ser272 and/or Thr334. When activated at

    Thr334, both p38 MAPK and MK2 translocate to the cytoplasm while still physically bound

    together using the CRM1-dependent mechanism, where both kinases activate their

    downstream substrates (Figure 1B). Phosphorylation at Thr222 within the activation loop is

    crucial for MK2-dependent activation of several target substrates, including enzymes,

    proteins that regulate cytoskeleton motility, mRNA binding proteins and regulators of cell

    cycle and apoptosis (Figure 2, reviewed in (Gaestel, 2006; Gaestel, 2013)}.

    The most extensively studied function of MK2 is the regulation of inflammation at the

    post-transcriptional level. The crucial role of MK2 in TNFα and interleukin-6 (IL-6)

    synthesis has been observed by phenotype analysis of MK2 knockout mice, which showed

    significantly reduced cytokines serum levels (Kotlyarov et al., 1999). Further studies have

    shown that MK2 modifies the stability and translation of TNFα and IL-6 mRNA through

    activation of AU-rich element-binding (ARE) proteins, such as tristetraproline (TTP),

    heterogeneous nuclear ribonucleoprotein A0 (hnRNP A0), polyA-binding protein-1 (PABP1)

    and human antigen-R (HuR) (Figure 2A). For example, MK2 phosphorylated TTP at Ser52

    and Ser178, two sites necessary for binding to 14-3-3 proteins (Chrestensen et al., 2004;

    Stoecklin et al., 2004). This phosphorylation abolished TTP function as TNFα suppressor by

    inhibiting ARE-mediated decay of TNFα mRNA and allowing efficient translation via

    subcellular translocation of the mRNA from P-bodies (Sandler and Stoecklin, 2008). TTP

    phosphorylation by MK2 also increased TTP protein expression via exclusion from

    proteasomal degradation and cytoplasmic retention (Deleault et al., 2008). Furthermore,

    MK2-dependent phosphorylation of TTP changed its affinity for ARE binding.

    Phosphorylated TTP had shown 10-fold lower Kd for ARE mRNA, and this led to the

    replacement of TTP from AREs by HuR, a protein known to stabilize short-lived mRNAs

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    7

    (Tiedje et al., 2012). These complex mechanisms of post-transcriptional regulation of

    cytokines synthesis via MK2-dependent phosphorylation of RNA binding proteins (RBPs)

    have been reviewed in several excellent reviews (Cargnello and Roux, 2011; Gaestel, 2013).

    The above-mentioned RBPs have been also reported to stabilize short-life mRNAs coding for

    cyclooxygenase 2 (COX-2), IL-8 and vascular endothelial growth factor (VEGF) (Miyata et

    al., 2013; Suswam et al., 2008). As the function of the RBPs is modified by MK2, but at the

    same time the substrate spectrum of MK2 is significantly smaller than that of p38 MAPK,

    MK2 has emerged as an attractive anti-inflammatory target.

    Although this review covers predominantly MK2, it is important to briefly reference

    MK3 (McLaughlin et al., 1996; Sithanandam et al., 1996). The expression levels of this

    kinase are much lower than MK2 (Ronkina et al., 2007), however the high structural identity

    and nearly the same substrate spectrum with MK2 (Cheng et al., 2010) imply similar

    functional behavior in the biological systems. Indeed, the C termini of MK3 contains NLS

    and NES signals rendering unphosphorylated MK3 in the nucleus and inducing translocation

    to the cytoplasm upon p38 MAPK-dependent phosphorylation. Furthermore, MK3 was

    involved in post-transcriptional regulation of ARE-containing mRNAs described for MK2

    (Figure 2), indicating that MK3 could control cytokines biosynthesis in addition to MK2

    (Ronkina et al., 2008). Indeed, MK2/3 double knockout mice showed higher reduction of

    lipopolysaccharide (LPS)-induced TNFα production than MK2 knockout animals (Ronkina et

    al., 2007). Intriguingly, functional differences between MK2 and MK3 have been shown in a

    similar system. Ehlting et al demonstrated that in LPS-stimulated macrophages, MK2

    regulates expression of interferon β by preventing MK3 from negative regulation of NFκB

    and IRF3 signaling (Ehlting et al., 2011).

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    8

    The role of MK2 in neuroinflammation

    Neurodegenerative conditions such as multiple sclerosis, Parkinson’s and Alzheimer’s

    diseases are associated with chronic neuroinflammation, which becomes problematic when

    unresolved. Furthermore, neuroinflammation worsens the progression and outcome of brain

    tumors, ischemic injury and epileptic seizures. Accumulating evidence points at the MK2 to

    be involved in the neuroinflammatory responses. MK2 is particularly enriched in microglia,

    the resident macrophages of the brain, and has been found to influence neurotoxicity through

    MK2-dependent activation of these immune cells (Culbert et al., 2006). Microglia from

    MK2-deficient (MK2-/-) mice showed significant inhibition of cytokine release upon LPS and

    interferon-γ (IFN-γ) stimulation, which abolished neurotoxicity in co-culture with neurons.

    Importantly, in the transgenic mouse model of Alzheimer’s disease, elevated activation and

    expression of MK2 correlated with amyloid-β deposition, microglial activation and cytokine

    up-regulation (Culbert et al., 2006). Further supporting the role of MK2 in microglial

    inflammation, Bachstetter et al demonstrated that anti-neuroinflammatory efficacy of the p38

    MAPK inhibitor MW01-2-069A-SRM is associated with decreased phosphorylation state of

    MK2 (Bachstetter et al., 2011). Similarly, in primary neuron-glia co-cultures, the

    dopaminergic neurons from MK2-deficient mice were significantly more resistant to LPS-

    induced neurotoxicity compared to cells from wild-type mice. This neuroprotection in MK2-

    deficient cultures was associated with reduced production of TNFα, IL-6 and nitric oxide.

    Furthermore, in the MPTP mouse model of Parkinson’s disease, MK2-deficient mice showed

    reduced neuroinflammation and less degeneration of dopaminergic neurons in substantia

    nigra (Thomas et al., 2008b).

    MK2 depletion also protected rodent brains from ischemic injury. MK2-deficient

    mice subjected to focal ischemia showed markedly reduced infarct size after transient and

    permanent ischemia as well as attenuation of neurological deficits. Biochemical analysis

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    9

    revealed decrease in IL-1β, but interestingly not TNFα production (Wang et al., 2002).

    Similarly, the inflammatory response after spinal cord injury (SCI) contributes to secondary

    tissue damage and functional loss. Microarray analysis of spinal cord tissues taken at the peak

    of the inflammatory response to SCI showed increased expression of phosphorylated MK2 in

    microglia and also in neurons and astrocytes. Locomotor recovery was significantly improved

    in MK2-deficient animals and was associated with reduced neuronal and myelin loss,

    decreased expression of pro-inflammatory cytokines and protein nitrosylation (Ghasemlou et

    al., 2010).

    Finally, MK2 has been reported to regulate synaptic plasticity that underlies the

    ability to learn and remember. Synaptic plasticity is stimulated by membrane depolarization

    and MK2 mRNA levels were described to be inducible by depolarization or after kainic acid

    induced seizures in the hippocampus (Vician et al., 2004). Depolarisation by potassium

    chloride or increasing cellular cAMP by forskolin treatment also led to elevated levels of

    MK2 expression. This induction of MK2 was characteristic of neuronal cells, and was absent

    in fibroblasts, macrophages and kidney cells. In vivo, induction of status epilepticus distinctly

    reduced neurodegeneration in MK2-deficient mice compared to wild-type mice (Thomas et

    al., 2008a). However, the down-stream substrates and detailed molecular mechanism of

    MK2-regulated neuroinflammation and neurodegeneration remain to be elucidated.

    MK2-dependent regulation of actin remodeling and cell migration

    Hsp27 (also known as Hspb1) is a member of the human small heat shock protein family

    characterized by highly conserved α-crystalline domain. It is generally recognized to act as

    molecular chaperone that can sequester damaged proteins and prevent their aggregation. This

    property of a molecular sponge is regulated via phosphorylation on three serine residues,

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    10

    namely Ser15, Ser78 and Ser82. Several kinases have been identified to phosphorylate Hsp27

    in vitro, however in cell culture and in vivo experiments the p38 MAPK-MK2 axis is the

    dominant kinase of Hsp27 phosphorylation (reviewed in (Kostenko and Moens, 2009)). This

    phosphorylation is a reversible process in response to variety of stimuli including mitogens,

    inflammatory cytokines IL-1β and TNFα, hydrogen peroxide and other oxidants.

    Unphosphorylated Hsp27 forms large multimers presenting chaperone functions, whereas

    phosphorylation of Hsp27 results in complex dissociation and loss of chaperoning activity.

    Hsp27-deficient mice are fertile without any obvious abnormalities (Crowe et al.,

    2013; Huang et al., 2007) and provide invaluable insight into the in vivo function of this

    chaperone. Hsp27 was found not essential for the embryonic development under

    physiological conditions, by was important, together with other Hsp proteins for tissue

    maintenance under stress conditions, where it provided cytoprotective and anti-apoptotic

    effects (Huang et al., 2007). Furthermore, Hsp27-deficient fibroblast showed increased

    expression of IL-6, but reduced entry into S-phase and increased expression of cyclin-

    dependent kinase inbitors p27kip1 and p21waf1, resulting in decreased proliferation. There was

    also a significant impairment in wound healing in Hsp27-deficient mice associated with

    collagen deposition, increased inflammation and augmented neutrophil infiltration into the

    wounds (Crowe et al., 2013). While hardly expressed in non-transformed cells, Hsp27 is

    abundantly expressed in cancer cells and has been investigated as a prognostic marker

    (Castro et al., 2012; Ischia and So, 2013). Furthermore, it accumulates in the cancer cells

    during chemo- and radiation therapy and thus it is strongly associated with resistance to

    anticancer therapy. The most prominent functions of Hsp27 are the control of apoptosis and

    cell migration through regulation of actin remodeling (Figure 2B).

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    11

    Actin remodeling requires the actin filament assembly and disassembly as well as the

    interaction between actin filaments and myosin thick filaments. Hsp27 is an actin filament

    capping protein that normally inhibits actin polymerization in its non-phosphorylated form.

    MK2-mediated phosphorylation of Hsp27 releases this inhibition and promotes actin

    polymerization and cell migration (Figure 2B). Disruption of balance between Hsp27

    phosphorylation and dephosphorylation states leads to either insufficient actin filaments,

    which prevents forward movements, or excessive amount of actin filaments that also inhibits

    cell movement by impeding the release of focal adhesion. Therefore, coordinated regulation

    of actin filament assembly through MK2-mediated Hsp27 phosphorylation is a critical

    component of actin filament dynamics that regulates cell migration. As such, MK2-/- mouse

    embryonic fibroblasts (MEFs) and smooth muscle cells showed reduced migration

    (Kotlyarov et al., 2002), while MK2-/- neutrophils showed a partial loss of directionality but

    higher migration speed (Hannigan et al., 2001).

    Cell migration is one of the essential biological processes in cancer metastasis. In

    prostate cancer model, both MK2 and Hsp27 were necessary for transforming growth factor β

    (TGFβ)-mediated cell invasion as transient transfection of dominant-negative MK2 or mutant

    Hsp27 each blocked TGFβ-induced increase in matrix metalloproteinase 2 (MMP-2) activity

    and consequent cell invasion (Xu et al., 2006). Hsp27 attenuation reversed epithelial-to-

    mesenchymal transition (EMT), an important determinant of prostate cancer metastasis, and

    decreased cell-migration, invasion and matrix metalloproteinase activity. Mechanistically,

    silencing Hsp27 was associated with decreased IL-6-dependent STAT3 phosphorylation,

    nuclear translocation and STAT3 binding to the Twist promoter. Importantly, Hsp27

    inhibition using antisense therapy decreased number of circulating tumor cells in patients

    with metastatic castration-resistant prostate cancer in a phase I clinical trials (Shiota et al.,

    2013). Immunohistochemistry of samples from 553 prostate cancer patients revealed that

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    12

    expression of Hsp27 is a reliable predictive biomarker of aggressive prostate cancer with

    poor clinical outcome (Cornford et al., 2000; Foster et al., 2009).

    Similarly, MK2 has been defined as a down-stream effector of p38 MAPK associated

    with MMP-2/9 activation in bladder cancer invasion (Kumar et al., 2010) and silencing

    Hsp27 using siRNA knockdown decreased metastatic behavior, invasion and migration in

    head and neck squamous cancer cells (HNSCC) (Zhu et al., 2010). The HNSCC study

    reported that the Hsp27 mRNA and protein levels are 22.4- and 25-fold higher, respectively,

    in metastatic UM-SCC-22B cell line when compared to primary UM-SCC-22A cells.

    Furthermore, Hsp27 gene was found to be overexpressed in a large fraction of the metastatic

    breast cancer patients. Depletion of Hsp27 in human breast cancer cell line MDA-

    MB231/B02 reduced cell migration and invasion and this attenuation of invasion correlated

    in vivo with a decreased ability of breast cancer cells to metastasize and grow in the skeleton

    (Gibert et al., 2012). In another comprehensive study, down-regulation of Hsp27 in MDA-

    MB-436-A breast cancer cells induced long-term dormancy in vivo (Straume et al., 2012).

    Only 4 of 30 Hsp27 knockdown xenograft tumor initiated growth after 70 days, which also

    correlated with a regain of Hsp27 protein expression. Importantly, no tumors escaped from

    dormancy without Hsp27 expression. Clinically, strong Hsp27 expression was associated

    with markers of aggressive tumor and decreased patient survival with breast cancer and

    melanoma.

    The MK2-dependent cell migration involves not only the direct p38 MAPK-MK2-

    Hsp27 phosphorylation axis. Chang et al showed that SUMOylation of MK2 at lysine339

    affects actin filament reorganization and cell migration (Chang et al., 2011). Loss of the MK2

    SUMOylation site increased MK2 kinase activity and prolonged Hsp27 phosphorylation,

    enhancing its effects on actin-filament-dependent events. Furthermore, MK2 phosphorylates

    LIM-kinase 1 (LIMK1) that in turn then phosphorylates cofilin, a protein that depolymerizes

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    13

    actin filaments. This activity of cofilin is reversibly regulated by phosphorylation and

    dephosphorylation of Ser3 residue, with the phosphorylated form being inactive. LIMK1 is

    responsible for phosphorylation of this site and can thereby inactivate cofilin and regulate

    stimulus-induced actin reorganization. The signaling pathway composed of p38 MAPK -

    MK2 - LIMK1 has also been identified critical for VEGF-induced stress fiber formation, cell

    migration and tubule formation (Kobayashi et al., 2006).

    In addition to its role in actin remodeling, Hsp27 is required for COX2 mRNA

    stabilization by the p38 MAPK – MK2 signaling cascade (Lasa et al., 2000) and also for IL-

    1β and TNFα-induced activation of TAK1 - p38 MAPK - MK2 and expression of

    inflammatory mediators including COX2, IL-6 and IL-8 (Alford et al., 2007) Interestingly,

    Hsp27 was found to affect upstream signaling cascades. Hsp27 deletion down-regulated p38

    MAPK - MK2 activation by IL-1β, resulting in COX-2 and IL-6 mRNA destabilization. This

    suggests that, at molecular level, Hsp27 contributes to the expression of pro-inflammatory

    mediators via regulation of the p38 MAPK - MK2-mediated mRNA stabilization.

    MK2 - Hsp27 axis in the brain

    In non-malignant diseases, Hsp27 overexpression has neuroprotective function. For example,

    it has been identified as an immediately secreted biomarker for ongoing ischemia that offers

    long-lasting neuroprotection via physical association with ASK1 and consequent inhibition of

    ASK1 - MKK4 - JNK pathway. The inhibition of this kinase cascade protected against

    progression of ischemic neuronal death (Stetler et al., 2008). Mechanistically, Hsp27 requires

    protein kinase D (PKD) mediated phosphorylation for its suppression of ASK1 cell death

    signaling (Stetler et al., 2012). Although several kinases have been shown to phosphorylate

    Hsp27 at Ser15, Ser78 and Ser82 in a context dependent manner, only PKD inhibitor

    CID755673 was able to block Hsp27 phosphorylation at Ser15 and Ser82 following oxygen

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    14

    and glucose deprivation (model of ischemia-like conditions in vitro). In further support,

    pharmacological inhibition of PKD, shRNA knockdown or overexpression of dominant

    negative PKD mutant significantly blocked the phospho-Hsp27-mediated neuroprotection

    against neuronal injury. Intriguing discovery of this study is that phosphorylation of Hsp27,

    that leads to dissociation of the Hsp27 oligomers and usually presents mechanism of Hsp27

    inactivation, had beneficial effects in the context of neuronal injury as it inhibited cell death

    via targeting mitochondrial signaling. The role of MK2 in this process has not been

    evaluated, but given that MK2 phosphorylates Hsp27 at Ser15, it is plausible to speculate that

    MK2 activity would be required for phospho-Hsp27-dependent neuroprotection. However,

    this is contradicting the neuroprotective phenotype observed in MK2 deficient animals

    through attenuation of the neuroinflammatory response to the injury (discussed above). One

    explanation could be that the loss of MK2-dependent Hsp27 phosphorylation at Ser15 is

    counterbalanced by activated PKD and thus the net effect of MK2 inhibition is

    neuroprotection. However, the exact role of MK2 in the neuroprotection remains to be

    elucidated.

    Aberrant expression and phosphorylation of Hsp27 has been implicated in some types

    of brain tumors. In gliomas, brain tumors derived from glial cells, the expression of Hsp27

    correlates with the degree of malignancy and highest Hsp27 expression was found in

    glioblastomas, the most aggressive Grade IV gliomas (Castro et al., 2012; Zhang et al., 2003).

    In addition, 4.7-fold increase in Hsp27 mRNA expression was found in hypoxic glioma

    regions, when compared to normoxic region in the in vivo approach of gene expression

    analysis. This indicates that Hsp27 could regulate hypoxia influence on biological function in

    solid tumors (Marotta et al., 2011). Similar to other cancers, MK2 - Hsp27 pathway has been

    implicated to drive glioma cell migration. In particular, secreted protein acidic and rich in

    cysteine (SPARC), a matricellular protein that negatively regulates cell proliferation while

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    15

    promoting migration and invasion, has been shown to activate p38 MAPK - MK2 pathway,

    leading to phosphorylation (thus inactivation) of Hsp27. Pre-treatment of SPARC-expressing

    glioma cells with Hsp27 siRNA prevented SPARC-induced migration and invasion

    (Golembieski et al., 2008). Furthermore, inhibition of Hsp27 phosphorylation alone, or in

    combination with pAkt inhibitor (for PTEN-null glioblastoma tumors) inhibited SPARC-

    induced invasion of brain cancer cells (Alam et al., 2013; Schultz et al., 2012). Phorbol-12-

    myristate-13-acetate (PMA)-induced migration of A172 glioma cells also activated p38

    MAPK and induced lamellipodia. Upon PMA stimulation, both unphosphorylated and

    phosphorylated Hsp27 were translocated to lamellipodia and the cell migration was abolished

    with p38 MAPK or Hsp27 knock-down (Nomura et al., 2007). As MK2 links p38 MAPK to

    Hsp27 phosphorylation, it is likely that MK2 deletion or inhibition would result in anti-

    migratory phenotype.

    Finally, phosphorylation status of Hsp27 was shown to have a switching role in the

    IL-1β induced IL-6 synthesis in C6 glioma cells. IL-1β induced IL-6 release in C6 glioma

    cells was significantly enhanced when C6 glioma cells were transfected with

    unphosphorylated Hsp27 and markedly suppressed when transfected with phosphorylated

    Hsp27 (Tanabe et al., 2010). However, the fact that chaperoning activity of unphosphorylated

    Hsp27 is required for IL-6 production contradicts the anti-IL6 efficacy of MK2 inhibitors that

    actually increase levels of unphosphorylated Hsp27. Thus, it can be hypothesized that anti-

    IL6 efficacy of MK2 inhibitors does not involve Hsp27, but another mechanisms such as

    activation of RNA-binding proteins that regulate IL-6 mRNA stability and translation.

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    16

    The involvement of MK2 pathway in cell cycle regulation

    Eukaryotic cells are in constant attack by internal or external sources causing base and DNA

    damage (Hoeijmakers, 2001). In order to repair damaged DNA and ensure that the genomic

    integrity is maintained during replication, cells undergo three cell cycle checkpoints, namely

    G1/S, intra-S and G2/M, before entering mitosis (Harper and Elledge, 2007; Kastan and

    Bartek, 2004). These checkpoints involve complex signaling mechanisms that provide an

    opportunity for the cells not only to repair their damaged DNA but also to exclude ones with

    extensive genotoxic insult (Abraham, 2001; Fridman and Lowe, 2003; Kastan and Bartek,

    2004; Sancar et al., 2004; Shiloh, 2003; Vousden and Lu, 2002). This process ensures that

    cells replicate their DNA successfully and that the integrity is sustained in daughter cells.

    Phosphatidylinositol-3-OH kinase-like kinases ataxia telangiectasia mutated (ATM)

    and ataxia telangiectasia and Rad3 related (ATR) are two major signaling networks that are

    activated in response to DNA damage (Figure 3) (Bakkenist and Kastan, 2003; Falck et al.,

    2005; Jazayeri et al., 2006). By activating checkpoint kinases 1 and 2 (Chk1/2) (Ahn et al.,

    2000; Culbert et al., 2006; Griffin et al., 2006; Kumagai et al., 2004; Liu et al., 2000;

    Schwarz et al., 2003) respectively, ATR and ATM control the G1/S, intra-S and G2/M cell

    cycle arrest for DNA repair (Berg et al., 2005; Chaturvedi et al., 1999; Chehab et al., 2000;

    Matsuoka et al., 1998; Sanchez et al., 1997). In addition, p38 MAPK-MK2 pathway,

    downstream of ATM and ATR, is activated in response to DNA damage caused by

    chemotherapy-induced G2/M arrest (Manke et al., 2005; Reinhardt et al., 2007). Mechanistic

    studies have further elucidated that a family of phosphatases, namely CDC25A, CDC25B and

    CDC25C function to activate specific CDK/cyclin complexes that in turn allow the

    progression of the cell cycle (Donzelli and Draetta, 2003; Hoffmann et al., 1994; Lammer et

    al., 1998; Peng et al., 1997). The checkpoint effector kinases, Chk1, Chk2 and MK2 have

    been described to inactivate CDC25 phosphatases causing CDK/cyclin inactivation and cell

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    17

    cycle arrest (Chen et al., 2003; Falck et al., 2001; Furnari et al., 1997; Manke et al., 2005;

    Reinhardt and Yaffe, 2009).

    To date, most studies have focused solely on p38 MAPK in cell cycle control and

    investigation of down-stream targets in this process is missing. p38 MAPK has been found to

    be activated in response to DNA damage or high osmotic shock causing G1/S and G2/M arrest

    (Bulavin et al., 2001; Dmitrieva et al., 2002; Hirose et al., 2003; Mikhailov et al., 2004;

    Pedraza-Alva et al., 2006). The p38 MAPK controls G1/S checkpoint by directly activating

    tumor suppressor p53 (Kishi et al., 2001; Takekawa et al., 2000) or stabilizing p21 mRNA

    (Lafarga et al., 2009). G2/M phase arrest is achieved by p38 MAPK phosphorylation-

    dependent inactivation of CDC25B/C (Ancrile et al., 2007; Bulavin et al., 2001; Hirose et al.,

    2003; Mikhailov et al., 2005). Biochemical phosphorylation studies revealed that p38 MAPK

    phosphorylated CDC25B at Ser323 and Ser375, whereas CDC25C is phosphorylated at the

    highly homologous Ser216 residue. Phosphorylation of these sites was required for binding to

    14-3-3 proteins and degradation (Bulavin et al., 2001). Lemaire et al extended these

    phosphorylation patterns of CDC25s inactivation by showing that p38 MAPK also

    phosphorylates CDC25B on Ser249 and that MK2 is able to phosphorylate CDC25B on

    multiple sites, including Ser169, Ser323, Ser353 and Ser375 (Lemaire et al., 2006).

    Moreover, UV irradiation caused G1/S and G2/M arrest in MK2 proficient U2OS cells, but

    not in the MK2 depleted ones. The abrogation of the cell cycle checkpoints in MK2-deficient

    cells forced cells to progress to mitosis with unrepaired DNA, which resulted in cell death as

    a consequence of mitotic catastrophe. Thus, inhibition of MK2 increased sensitivity to UV

    induced DNA damage. In mechanistic detail, MK2 was shown to directly phosphorylate

    CDC25B on Ser323 resulting in its binding to 14-3-3 and inactivation. This phosphatase

    inactivation in turn resulted in cell cycle arrest (Manke et al., 2005).

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    18

    The role of MK2 in checkpoint signaling has been reinforced by Reinhardt et al.

    (Reinhardt et al., 2007) who demonstrated that p53-deficient cells rely on MK2 for cell cycle

    arrest and survival after DNA damage caused by cisplatin and doxorubicin. Furthermore,

    MK2 depletion in p53-deficient MEFs, but not in p53-wild type cells, caused abrogation of

    the CDC25A mediated S phase arrest after cisplatin treatment as well as the CDC25B

    mediated G2/M arrest following doxorubicin exposure, resulting in enhanced anti-

    proliferative effects and sensitization to the chemotherapeutics (Reinhardt et al., 2007). The

    activation of MK2 after the drug induced DNA damage required activity of upstream ATM

    and ATR kinases, however occurred independently of Chk1. Conversely, Chk1 activation

    was independent of MK2 activity. This indicates that the ATM/ATR - p38 MAPK - MK2

    pathway functions in parallel with ATR - Chk1 pathway in response to DNA damage by

    chemotherapeutics. In a further study, Reinhardt et al. (Reinhardt et al., 2010) illustrated that

    in p53-deficient cells Chk1 mediates an early nuclear G2/M checkpoint, whereas MK2-

    mediated a late cytoplasmic checkpoint. This was achieved by translocation of the activated

    p38 MAPK - MK2 complex from the nucleus to the cytoplasm where MK2 phosphorylated

    hnRNP A0 and PARN to ultimately stabilize Gadd45α mRNA. Gadd45α in turn maintained

    the MK2 activity in the cytoplasm and hence cytoplasmic sequestration of CDC25B/C

    preventing CDK1 activation and mitotic entry until DNA damage was repaired.

    Another mode by which MK2 may interfere with the cell cycle is through modulating

    the activity of the tumor suppressor p53, a target of p38 MAPK essential for cell cycle

    regulation at G1/S and entry into apoptosis (Bulavin et al., 1999). MK2 was found to increase

    the degradation of p53 through phosphorylation of HDM2, a p53-interacting ubiquitin ligase.

    It was therefore proposed that MK2 dampens the extent and duration of p53 activity and

    contributes to the fine-tuning of the DNA damage response. The consequence of MK2

    depletion was increased p53 protein levels and improved sensitivity of MK2-/- MEF cells to

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    19

    UV induced apoptosis compared to wild type cells (Weber et al., 2005). In further support, a

    study published in 2012 demonstrated MK2 as a repressor of p53-driven apoptosis (Hopker et

    al., 2012). MK2-dependent phosphorylation of apoptosis-antagonising transcription factor

    (AATF) caused dissociation of p-AATF from cytoplasmic myosin-regulatory light chain 3

    (MRLC3) and subsequent nuclear translocation (Figure 2C). In the nucleus, p-AATF binds to

    the PUMA, BAX, BAK promoter regions to repress p53-driven expression of these pro-

    apoptotic proteins. However, nuclear AATF did not restrict the expression of the cell-cycle

    regulating genes CDKN1A, Gadd45α and RPRM. The net result was a selective repression of

    p53-driven apoptosis in response to adriamycin. Thus, inhibiting MK2 pathway could be

    explored in chemotherapy-sensitizing approaches to treat both p53-deficient and p53-

    proficient cancers.

    However, two studies have called MK2 involvement in checkpoint control into

    question. Firstly, Xiao et al. found that Chk1 knockdown, but not MK2 or Chk2, was

    sufficient to abrogate S phase and G2/M arrest after chemotherapy-induced DNA damage in

    HeLa and H1299 cells. Intriguingly, simultaneous depletion of Chk1 and MK2 partially

    reversed the checkpoint abrogation observed with Chk1 knockdown alone. While depletion

    of Chk1 increased CDC25A levels, loss of MK2 destabilized CDC25A protein. Hence, it was

    suggested that MK2 prevents Chk1-induced degradation of CDC25A, which is required for

    the checkpoint abrogation and cell cycle progression (Xiao et al., 2006). Secondly, Phong et

    al. highlighted that p38 MAPK - MK2 pathway is not required for G2/M DNA damage

    checkpoint control but rather plays an important cytoprotective role through regulation of

    anti-apoptotic and survival pathways to allow cells to recover from DNA damage (Phong et

    al., 2010). Cancer cells were still able to undergo a G2/M arrest in response to doxorubicin,

    methyl methanesulfonate or UV-induced DNA damage despite p38 MAPK or MK2

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    20

    inhibition, but this inhibition triggered dramatic apoptosis in a p53-independent manner

    associated with decreased levels of anti-apoptotic Bcl-2 proteins.

    In addition to its role in checkpoint control and mitotic entry, p38 MAPK - MK2

    pathway has been suggested to regulate mitotic progression as well, although in less details.

    The discovery that p38 MAPK knockdown strongly inhibited HeLa cell proliferation,

    instigated examination of the underlying mechanism (Fan et al., 2005). In the absence of

    p38α MAPK, cells underwent a G2/M arrest and exhibited marked defects in bipolar spindle

    formation and chromosome alignment. Intriguingly, this essential function of p38 MAPK for

    proper mitotic progression does not require its kinase activity. Likewise, loss of p38γ MAPK

    isoform in HeLa cells resulted in multipolar spindle formation and chromosome

    misalignment, which caused M phase arrest. As a result, the p38 MAPK-depleted cells died

    at the mitotic arrest or soon after abnormal exit from M phase via caspase-dependent

    apoptosis. In addition, activated p38 MAPK was found to localize on kinetochores and

    spindle poles during mitosis, which is crucial for the normal kinetochore localization of polo-

    like kinase 1 (Plk1) that regulates centrosome maturation, sister chromatid segregation and

    cytokinesis (Barr et al., 2004; Kukkonen-Macchi et al., 2011). The expression of a mutant

    Plk1 or depletion of Plk1 caused spindle pole defects, accumulation of cells at M phase and

    massive cell death (Bu et al., 2008; Guan et al., 2005; Reagan-Shaw and Ahmad, 2005). Tang

    et al described that MK2 co-localizes with activated p38 MAPK and Plk1 at spindle poles

    where it phosphorylates Plk1 at Ser326 to promote normal mitotic progression (Figure 2D)

    (Tang et al., 2008). Importantly, the same study reported that MK2 acts as a Plk1 kinase and

    that MK2 knockdown resulted in mitotic arrest. In summary, MK2 could function not only as

    a checkpoint kinase that controls mitotic entry, but appears to be involved in mitotic

    progression.

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    21

    A new, less investigated, role for MK2 was demonstrated by Chen et al (Chen et al.,

    2000). This work showed that Ras activated the p38 MAPK pathway, including the upstream

    kinase MKK3 and down-stream kinases MK2 and MK5 (also known as PRAK). Each of

    these kinases, when activated by Ras, was able to inhibit Ras-induced proliferation in tumor

    cells harboring endogenous Ras mutations. Mechanistically, the negative feedback of MK2,

    which completely blocked Ras proliferative signaling was achieved via inhibition of Ras-

    induced JNK activation. A follow-up study (Kobayashi et al., 2012) demonstrated that MK2

    knockdown increases Ras transformation in MEFs and enhances tumorigenesis in vivo.

    However, opposite role for MK2 was demonstrated in human colon cancer cells, where MK2

    depletion decreased the tumor growth. It was suggested that the differing effects of MK2 are

    related to its effects on reactive oxygen species (ROS), as in MEFs MK2 decreased levels of

    ROS, but increased its production in human colon cancer cells.

    Finally, adding more to the puzzle of MK2-controlled cell survival, MK2 knockdown

    has been reported to protect cells from DNA damage-induced cell death (Köpper et al.,

    2013). Mice deficient for MK2 displayed decreased apoptosis in the skin upon UV

    irradiation, accompanied with reduced H2AX phosphorylation. MK2 inhibition increased

    survival of gemcitabine-treated U2OS cells. In contrast, Chk1 knockdown or

    pharmacological inhibition strongly enhanced H2AX phosphorylation in gemcitabine-treated

    cells, suggesting that loss of MK2 in tumors is likely to constitute resistance to Chk1

    inhibition.

    The therapeutic potential of targeting MK2

    Small-molecule MK2 inhibitors are “tough nut to crack” (Schlapbach and Huppertz, 2009)

    because of several challenges associated with their development. Firstly, MK2 crystal

    structures revealed that the ATP pocket of MK2 is deep and narrow (Anderson et al., 2007;

    Hillig et al., 2007), allowing only small planar molecules to bind and restricting addition of

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    22

    side chains that define kinase selectivity. Secondly, low biochemical efficiency (BE) index of

    MK2 inhibitors hinders development of MK2 inhibitors into therapeutics. BE is defined as

    binding affinity/functional response ratio, which is equivalent to Ki/EC50 (Swinney and

    Anthony, 2011). Properties influencing BE are assay relevance, molecular properties of the

    drug, target engagement and the molecular mechanism of action; however given that all

    aspects are addressed properly in the early stages of drug discovery, BE is a good indicator of

    clinical success. Swinney et al showed that BE > 0.4 is a property of many approved

    medicines (Swinney, 2004). In other words, for the majority of successful drugs, the cellular

    EC50 values are no more than 2.5-fold higher than biochemical binding or inhibition Ki/IC50

    values. However, cellular efficacy data of MK2 inhibitors in disease-relevant assays are

    limited in the public domain and BE calculated from these published values are far below the

    0.4 threshold (Anderson et al., 2009; Kaptein et al., 2011; Mourey et al., 2010; Oubrie et al.,

    2012). It is believed that the low BE is caused by high affinity of un-phosphorylated

    (inactive) MK2 to ATP (Mourey et al., 2010). As many kinases have low ATP affinity in the

    inactive conformation, as opposed to the high ATP affinity in the active conformation, their

    respective inhibitors explore predominantly the inactive conformation; thus avoiding

    competition with the high intracellular ATP concentration. This advantage is not available for

    MK2 inhibitors and therefore higher inhibitor concentrations are required for cellular

    efficacy. As good BE enables efficacy at lower drug concentrations and increases the

    therapeutic index, there is low probability of clinical success for ATP-competitive MK2

    inhibitors. Nevertheless, Mourey et al demonstrated in vivo efficacy of a selective ATP-

    competitive MK2 inhibitor PF-3644022 despite its biochemical inefficiency (BE = 0.03). PF-

    3644022 reduced TNFα production and paw swelling in acute and chronic models of

    inflammation (Mourey et al., 2010). This data, supported by the normal phenotype of MK2

    knockout mice (Kotlyarov et al., 1999), suggests that MK2 is a viable target that requires

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    23

    non-ATP competitive or allosteric approach. Non-ATP competitive inhibitors have been

    reported by Merck (Huang et al., 2012; Qin et al., 2011) and it will be interesting to see how

    this class of compounds will progress through in vivo studies. Until then, we can only assume

    the outcomes of MK2 inhibition by analyzing efficacy of p38 MAPK inhibitors that inhibit

    MK2 activation or by analyzing efficacy of Chk1 or Wee1 inhibitors as these kinases have

    similar functions in cell cycle progression. Along these lines, Watterson et al. has recently

    demonstrated that anti-neuroinflammatory efficacy of BBB-permeable p38 MAPK inhibitors

    in the animal model of Alzheimer’s disease correlates with the inhibition of MK2 activity

    (Watterson et al., 2013).

    p38 MAPK, Chk1 and Wee1 inhibitors in cancer: Implication for MK2

    inhibitors

    By arresting cell cycle progression, cancer cells are capable of repairing the DNA damage

    caused by chemotherapeutic agents and escape apoptosis. The implication that MK2 activity

    is necessary for G2/M checkpoint arrest provides an exciting possibility for the use of MK2

    inhibitors as chemosensitizers. Indeed, MK2 depletion improved the effectiveness of

    chemotherapeutics in p53-deficient, but not in p53-proficient cells (Reinhardt et al., 2007).

    As most cancers are p53 mutated (Vogelstein et al., 2000), they can be selectively targeted

    with MK2 inhibition. This synthetic lethality is due to the fact that in p53-proficient cells, the

    cell cycle checkpoints are well maintained through p53 and Chk1 responses (Lam et al.,

    2004). On the other hand, the loss of p53 results in cells being entirely dependent on intra-S

    and G2/M checkpoints to maintain their genomic integrity in response to DNA damage.

    Consequently, the p38 MAPK - MK2 pathway becomes more crucial for cell survival after

    DNA damage. Thus, it can be hypothesized that inhibition of MK2 will deliver outcome

    similar to inhibition of Chk1 or Wee1, both being targets of clinical investigation (Patil et al.,

    2013). Importantly, MK2-depleted mice are viable (Hegen et al., 2006; Kotlyarov et al.,

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    24

    1999; Ronkina et al., 2007) in contrast to Chk1 and p38 MAPK knockout mice (Liu et al.,

    2000; Takai et al., 2000), indicating that MK2 inhibition would successfully target cancer

    cells like Chk1 and p38 MAPK inhibitors, but with fewer side effects.

    In brain cancer, the knowledge whether p38 MAPK - MK2 inhibition could improve

    chemotherapy sensitivity is limited to in vitro data using only pharmacological inhibitors of

    p38 MAPK. Hirose et al. reported that U87 glioblastoma cells underwent a p38 MAPK-

    dependent G2/M arrest in response to the DNA methylating agent temozolomide (TMZ)

    (Hirose et al., 2003). Pharmacological blocking of p38 MAPK activity with SB203580 or

    siRNA knockdown bypassed the G2/M checkpoint resulting in increased sensitivity to

    cytotoxic action of TMZ. Interestingly, the p38 MAPK and Chk1 were found to work

    cooperatively to activate G2/M arrest (Hirose et al., 2004; Llopis et al., 2012). Both pathways

    were shown to deactivate CDC25C in response to TMZ treatment resulting in CDK1

    inactivation and G2/M arrest in U87 glioblastoma cells. Moreover, pharmacological inhibition

    of both pathways did not lead to greater bypass of TMZ induced G2/M arrest or enhanced

    cytotoxicity than inhibition of either pathway alone. However, Chk1 was found to be crucial

    for both the initiation and maintenance of the TMZ induced G2/M arrest, whereas p38 MAPK

    appeared to be important only for initiation. Further supporting this approach to brain cancer

    therapy, inhibition of ATM/ATR (Eich et al., 2013), Chk1 (Hirose et al., 2001) or Wee1 (Mir

    et al., 2010) potentiated TMZ efficacy in brain cancer in vivo models. In addition, Wee1

    kinase was demonstrated as a major regulator of the G2 checkpoint in glioblastoma (Mir et

    al., 2010) and Wee1 inhibitor MK-1775 enhanced radiosensitivity in established glioma cell

    lines in vitro and in vivo, without modulating response in normal human astrocytes (Sarcar et

    al., 2011).

    As discussed above, the p38 MAPK-MK2 pathway appears to play a role in mitosis

    progression through Plk1 activation. Therefore, it is possible that inhibition of MK2 could

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    25

    deliver outcomes similar to Plk1 inhibition, such as defects in bipolar spindle formation and

    cytokinesis, growth inhibition and induction of apoptosis. The anti-cancer efficacy of Plk1

    inhibition, without significant toxicity in non-transformed cells, has been demonstrated in

    oesophageal, prostate and brain cancer models (Bu et al., 2008; Guan et al., 2005; Liu and

    Erikson, 2003; Reagan-Shaw and Ahmad, 2005; Strebhardt, 2010; Tandle et al., 2013).

    Finally, based on the concept that many tumors exhibit high levels of replicative

    stress, the sole inhibition of Chk1 has been shown to effectively target melanoma cells with

    significant intrinsic DNA damage (Brooks et al., 2013). The cytotoxic effect of Chk1

    inhibitors resulted from inhibition of Chk1 activity in the S phase driving premature exit from

    S phase into an aberrant mitosis resulting in either failure of cytokinesis or cell death by an

    apoptotic mechanism. Likewise, Ferrao et al showed that inhibition of Chk1 effectively

    targeted lymphoma cells with MYC-driven intrinsic replicative stress but not normal B cells

    (Ferrao et al., 2012). Similarly, Wee1 inhibition by MK-1775 has been shown as a potent

    anticancer therapy approach independent of a genotoxic agent (Guertin et al., 2013).

    Concluding remarks

    Various functions of MK2 have been outlined herein that provide opportunities to intervene

    in the treatment of brain diseases (Figure 2). Current evidence suggests that MK2 contributes

    to neuroinflammation and plays a role in the pathophysiology of conditions such as

    Parkinson’s and Alzheimer’s disease. MK2 regulates the expression of various inflammatory

    cytokines and thus is emerging as a novel target for neuroinflammation-associated brain

    disorders. In addition, MK2-dependent regulation of Hsp27 has been shown to be crucial in

    actin remodeling and cell migration. While being more prominently expressed in cancer cells

    than normal cells, modulating Hsp27 activity appears to be an interesting strategy to reduce

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    26

    migration and metastasis of cancer. Indeed, inhibition of MK2 or Hsp27 reduced the

    migration and invasion of a number of cancers such as HNSCC and breast cancer. The role of

    MK2 in controlling cell cycle arrest in response to DNA damage much similar to Chk1 could

    have positive implications in cancer therapy. Improving chemotherapy effectiveness by

    inhibiting MK2 has been already demonstrated in in vitro studies. However, translation of

    these interesting in vitro data into the in vivo proof-of-principle stage is challenging. Many

    MK2 inhibitors have been developed, nevertheless, needing to compete with high

    intracellular ATP, their BE is below the threshold required for therapeutic efficacy. Focusing

    on brain disorders, another challenge for MK2 inhibitors to overcome is the blood-brain

    barrier permeability, as well as the ability to avoid excessive efflux by the P-glycoprotein

    transporter. Finally, it is important to note that the efficacy of MK2 inhibitors will also

    depend on other factors such as target engagement in the disease pathophysiology and/or the

    (in)ability of the targeted cells to bypass MK2 inhibition.

    Conflict of interest

    The authors declare no conflict of interest.

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    27

    Acknowledgements

    FMSG is supported by the University of Sydney Australian Postgraduate Award.

    Authorship Contributions.

    Wrote or contributed to the writing of the manuscript: Gurgis, Ziaziaris, Munoz.

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    28

    References

    Abraham RT (2001) Cell cycle checkpoint signaling through the ATM and ATR kinases.

    Genes Dev 15: 2177-2196.

    Ahn JY, Schwarz JK, Piwnica-Worms H and Canman CE (2000) Threonine 68

    phosphorylation by ataxia telangiectasia mutated is required for efficient activation of

    Chk2 in response to ionizing radiation. Cancer Res 60: 5934-5936.

    Alam R, Schultz CR, Golembieski WA, Poisson LM and Rempel SA (2013) PTEN

    suppresses SPARC-induced pMAPKAPK2 and inhibits SPARC-induced Ser78

    HSP27 phosphorylation in glioma. Neuro Oncol 15: 451-461.

    Alford KA, Glennie S, Turrell BR, Rawlinson L, Saklatvala J and Dean JLE (2007) Heat

    shock protein 27 functions in inflammatory gene expression and transforming growth

    factor-β-activated kinase-1 (TAK1)-mediated signaling. J Biol Chem 282: 6232-6241.

    Ancrile B, Lim K-H and Counter CM (2007) Oncogenic Ras-induced secretion of IL6 is

    required for tumorigenesis. Genes Dev 21: 1714-1719.

    Anderson DR, Meyers MJ, Kurumbail RG, Caspers N, Poda GI, Long SA, Pierce BS,

    Mahoney MW and Mourey RJ (2009) Benzothiophene inhibitors of MK2. Part 1:

    Structure–activity relationships, assessments of selectivity and cellular potency.

    Bioorg Med Chem Lett 19: 4878-4881.

    Anderson DR, Meyers MJ, Vernier WF, Mahoney MW, Kurumbail RG, Caspers N, Poda GI,

    Schindler JF, Reitz DB and Mourey RJ (2007) Pyrrolopyridine inhibitors of mitogen-

    activated protein kinase-activated protein kinase 2 (MK-2). J Med Chem 50: 2647-

    2654.

    Bachstetter A, Xing B, de Almeida L, Dimayuga E, Watterson DM and Van Eldik L (2011)

    Microglial p38alpha MAPK is a key regulator of proinflammatory cytokine up-

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    29

    regulation induced by toll-like receptor (TLR) ligands or beta-amyloid (Abeta). J

    Neuroinflammation 8: 79.

    Bakkenist CJ and Kastan MB (2003) DNA damage activates ATM through intermolecular

    autophosphorylation and dimer dissociation. Nature 421: 499-506.

    Barr FA, Sillje HH and Nigg EA (2004) Polo-like kinases and the orchestration of cell

    division. Nat Rev Mol Cell Biol 5: 429-440.

    Ben-Levy R, Hooper S, Wilson R, Paterson HF and Marshall CJ (1998) Nuclear export of the

    stress-activated protein kinase p38 mediated by its substrate MAPKAP kinase-2. Curr

    Biol 19: 1049-1057.

    Ben-Levy R, Leighton I, Doza Y, Attwood P, Morrice N, marshall C and Cohen P (1995)

    Identification of novel phosphorylation sites required for activation of MAPKAP

    kinase-2. EMBO J 14: 5920-5930.

    Berg BM, Godbout JP, Chen J, Kelley KW and Johnson RW (2005) {alpha}-Tocopherol and

    selenium facilitate recovery from lipopolysaccharide-induced sickness in aged mice. J

    Nutr 135: 1157-1163.

    Brooks K, Oakes V, Edwards B, Ranall M, Leo P, Pavey S, Pinder A, Beamish H,

    Mukhopadhyay P, Lambie D and Gabrielli B (2013) A potent Chk1 inhibitor is

    selectively cytotoxic in melanomas with high levels of replicative stress. Oncogene

    32: 788-796.

    Bu Y, Yang Z, Li Q and Song F (2008) Silencing of polo-like kinase (Plk) 1 via siRNA

    causes inhibition of growth and induction of apoptosis in human esophageal cancer

    cells. Oncology 74: 198-206.

    Bulavin DV, Higashimoto Y, Popoff IJ, Gaarde WA, Basrur V, Potapova O, Appella E and

    Fornace AJ, Jr. (2001) Initiation of a G2/M checkpoint after ultraviolet radiation

    requires p38 kinase. Nature 411: 102-107.

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    30

    Bulavin DV, Saito S, Hollander MC, Sakaguchi K, Anderson CW, Appella E and Fornace

    AJ, Jr. (1999) Phosphorylation of human p53 by p38 kinase coordinates N-terminal

    phosphorylation and apoptosis in response to UV radiation. EMBO J 18: 6845-6854.

    Cargnello M and Roux PP (2011) Activation and function of the MAPKs and their substrates,

    the MAPK-activated protein kinases. Microbiol Mol Biol Rev 75: 50-83.

    Castro G, Cayado-Gutiérrez N, Moncalero V, Lima P, Angelis R, Chávez V, Cuello-Carrión

    FD and Ciocca D (2012) Hsp27 (HSPB1): a possible surrogate molecular marker for

    loss of heterozygosity (LOH) of chromosome 1p in oligodendrogliomas but not in

    astrocytomas. Cell Stress Chaperones 17: 779-790.

    Chang E, Heo K-S, Woo C-H, Lee H, Le N-T, Thomas TN, Fujiwara K and Abe J-i (2011)

    MK2 SUMOylation regulates actin filament remodeling and subsequent migration in

    endothelial cells by inhibiting MK2 kinase and HSP27 phosphorylation. Blood 117:

    2527-2537.

    Chaturvedi P, Eng WK, Zhu Y, Mattern MR, Mishra R, Hurle MR, Zhang X, Annan RS, Lu

    Q, Faucette LF, Scott GF, Li X, Carr SA, Johnson RK, Winkler JD and Zhou BB

    (1999) Mammalian Chk2 is a downstream effector of the ATM-dependent DNA

    damage checkpoint pathway. Oncogene 18: 4047-4054.

    Chehab NH, Malikzay A, Appel M and Halazonetis TD (2000) Chk2/hCds1 functions as a

    DNA damage checkpoint in G(1) by stabilizing p53. Genes Dev 14: 278-288.

    Chen F, Wang M, O'Connor JP, He M, Tripathi T and Harrison LE (2003) Phosphorylation

    of PPARgamma via active ERK1/2 leads to its physical association with p65 and

    inhibition of NF-kappabeta. J Cell Biochem 90: 732-744.

    Chen G, Hitomi M, Han J and Stacey DW (2000) The p38 pathway provides negative

    feedback for Ras proliferative signaling. J Biol Chem 275: 38973-38980.

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    31

    Cheng R, Felicetti B, Palan S, Toogood-Johnson I, Scheich C, Barker J, Whittaker M and

    Hesterkamp T (2010) High-resolution crystal structure of human Mapkap kinase 3 in

    complex with a high affinity ligand. Protein Sci 19: 168-173.

    Chrestensen CA, Schroeder MJ, Shabanowitz J, Hunt DF, Pelo JW, Worthington MT and

    Sturgill TW (2004) MAPKAP kinase 2 phosphorylates tristetraprolin on in vivo sites

    including Ser178, a site required for 14-3-3 binding. J Biol Chem 279: 10176-10184.

    Cornford PA, Dodson AR, Parsons KF, Desmond AD, Woolfenden A, Fordham M,

    Neoptolemos JP, Ke Y and Foster CS (2000) Heat shock protein expression

    independently predicts clinical outcome in prostate cancer. Cancer Res 60: 7099-

    7105.

    Crowe J, Aubareda A, McNamee K, Przybycien PM, Lu X, Williams RO, Bou-Gharios G,

    Saklatvala J and Dean JLE (2013) Heat shock protein B1-deficient mice display

    impaired wound healing. PLoS ONE 8: e77383.

    Culbert AA, Skaper SD, Howlett DR, Evans NA, Facci L, Soden PE, Seymour ZM, Guillot

    F, Gaestel M and Richardson JC (2006) MAPK-activated protein kinase 2 deficiency

    in microglia inhibits pro-inflammatory mediator release and resultant neurotoxicity.

    Relevance to neuroinflammation in a transgenic mouse model of Alzheimer disease. J

    Biol Chem 281: 23658 - 23667.

    Deleault KM, Skinner SJ and Brooks SA (2008) Tristetraprolin regulates TNF TNF-α mRNA

    stability via a proteasome dependent mechanism involving the combined action of the

    ERK and p38 pathways. Mol Immunol 45: 13-24.

    Dmitrieva NI, Bulavin DV, Fornace AJ, Jr. and Burg MB (2002) Rapid activation of G2/M

    checkpoint after hypertonic stress in renal inner medullary epithelial (IME) cells is

    protective and requires p38 kinase. Proc Natl Acad Sci USA 99: 184-189.

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    32

    Donzelli M and Draetta GF (2003) Regulating mammalian checkpoints through Cdc25

    inactivation. EMBO reports 4: 671-677.

    Ehlting C, Ronkina N, Böhmer O, Albrecht U, Bode KA, Lang KS, Kotlyarov A, Radzioch

    D, Gaestel M, Häussinger D and Bode JG (2011) Distinct functions of the mitogen-

    activated protein kinase-activated protein (MAPKAP) kinases MK2 and MK3: MK2

    mediates lipopolysaccharide-induced signal transducers and activators of transcription

    3 (STAT3) activation by preventing negative regulatory effects of MK3. J Biol Chem

    286: 24113-24124.

    Eich M, Roos WP, Nikolova T and Kaina B (2013) Contribution of ATM and ATR to the

    resistance of glioblastoma and malignant melanoma cells to the methylating

    anticancer drug temozolomide. Mol Cancer Ther 12: 2529-2540.

    Engel K, Kotlyarov A and Gaestel M (1998) Leptomycin B-sensitive nuclear export of

    MAPKAP kinase 2 is regulated by phosphorylation. EMBO J 17: 3363-3371.

    Falck J, Coates J and Jackson SP (2005) Conserved modes of recruitment of ATM, ATR and

    DNA-PKcs to sites of DNA damage. Nature 434: 605-611.

    Falck J, Mailand N, Syljuasen RG, Bartek J and Lukas J (2001) The ATM-Chk2-Cdc25A

    checkpoint pathway guards against radioresistant DNA synthesis. Nature 410: 842-

    847.

    Fan L, Yang X, Du J, Marshall M, Blanchard K and Ye X (2005) A novel role of p38 alpha

    MAPK in mitotic progression independent of its kinase activity. Cell Cycle 4: 1616-

    1624.

    Ferrao PT, Bukczynska EP, Johnstone RW and McArthur GA (2012) Efficacy of CHK

    inhibitors as single agents in MYC-driven lymphoma cells. Oncogene 31: 1661-1672.

    Foster CS, Dodson A, Ambroisine L, Fisher G, Moller H, Clark J, Attard G, De-bono J,

    Scardino P, Reuter V, Cooper C, Berney D and Cuzick J (2009) Hsp-27 expression at

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    33

    diagnosis predicts poor clinical outcome in prosttae cancer independent of ETS-gene

    rearrangement. Br J Cancer 101: 1137-1144.

    Fridman JS and Lowe SW (2003) Control of apoptosis by p53. Oncogene 22: 9030-9040.

    Furnari B, Rhind N and Russell P (1997) Cdc25 mitotic inducer targeted by chk1 DNA

    damage checkpoint kinase. Science 277: 1495-1497.

    Gaestel M (2006) MAPKAP kinases - MKs - two's company, three's a crowd. Nat Rev Mol

    Cell Biol 7: 120-130.

    Gaestel M (2013) What goes up must come down: molecular basis of MAPKAP kinase 2/3-

    dependent regulation of the inflammatory response and its inhibition. Biol Chem 394:

    1301-1315.

    Ghasemlou N, Lopez-Vales R, Lachance C, Thuraisingam T, Gaestel M, Radzioch D and

    David S (2010) Mitogen-activated protein kinase-activated protein kinase 2 (MK2)

    contributes to secondary damage after spinal cord injury. J Neurosci 30: 13750-

    13759.

    Gibert B, Eckel B, Gonin V, Goldschneider D, Fombonne J, Deux B, Mehlen P, Arrigo A,

    Clezardin P and Diaz-Latoud C (2012) Targeting heat shock protein 27 (HspBI)

    interferes with bone metastasis and tumour formation in vivo. Br J Cancer 107: 63-

    70.

    Golembieski WA, Thomas SL, Schultz CR, Yunker CK, McClung HM, Lemke N, Cazacu S,

    Barker T, Sage EH, Brodie C and Rempel SA (2008) HSP27 mediates SPARC-

    induced changes in glioma morphology, migration, and invasion. Glia 56: 1061-1075.

    Griffin WST, Liu L, Li Y, Mrak RE and Barger SW (2006) Interleukin-1 mediates Alzheimer

    and Lewy body pathologies. J Neuroinflammation 3: 5.

    Guan R, Tapang P, Leverson JD, Albert D, Giranda VL and Luo Y (2005) Small interfering

    RNA-mediated Polo-like kinase 1 depletion preferentially reduces the survival of p53-

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    34

    defective, oncogenic transformed cells and inhibits tumor growth in animals. Cancer

    Res 65: 2698-2704.

    Guertin AD, Li J, Liu Y, Hurd MS, Schuller AG, Long B, Hirsch HA, Feldman I, Benita Y,

    Toniatti C, Zawel L, Fawell SE, Gilliland DG and Shumway SD (2013) Preclinical

    Evaluation of the WEE1 Inhibitor MK-1775 as Single-Agent Anticancer Therapy.

    Mol Cancer Ther 12: 1442-1452.

    Hannigan MO, Zhan L, Ai Y, Kotlyarov A, Gaestel M and Huang C-K (2001) Abnormal

    Migration Phenotype of Mitogen-Activated Protein Kinase-Activated Protein Kinase

    2−/− Neutrophils in Zigmond Chambers Containing Formyl-Methionyl-Leucyl-

    Phenylalanine Gradients. J Immunol 167: 3953-3961.

    Harper JW and Elledge SJ (2007) The DNA damage response: ten years after. Mol Cell 28:

    739-745.

    Hegen M, Gaestel M, Nickerson-Nutter CL, Lin LL and Telliez JB (2006) MAPKAP kinase

    2-deficient mice are resistant to collagen-induced arthritis. J Immunol 177: 1913-

    1917.

    Hillig RC, Eberspaecher U, Monteclaro F, Huber M, Nguyen D, Mengel A, Muller-Tiemann

    B and Egner U (2007) Structural basis for a high affinity inhibitor bound to protein

    kinase MK2. J Mol Biol 369: 735-745.

    Hirose Y, Berger MS and Pieper RO (2001) Abrogation of the Chk1-mediated G(2)

    checkpoint pathway potentiates temozolomide-induced toxicity in a p53-independent

    manner in human glioblastoma cells. Cancer Res 61: 5843-5849.

    Hirose Y, Katayama M, Berger MS and Pieper RO (2004) Cooperative function of Chk1 and

    p38 pathways in activating G2 arrest following exposure to temozolomide. J

    Neurosurgery 100: 1060-1065.

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    35

    Hirose Y, Katayama M, Stokoe D, Haas-Kogan DA, Berger MS and Pieper RO (2003) The

    p38 Mitogen-Activated Protein Kinase Pathway Links the DNA Mismatch Repair

    System to the G2 Checkpoint and to Resistance to Chemotherapeutic DNA-

    Methylating Agents. Mol Cell Biol 23: 8306-8315.

    Hoeijmakers JH (2001) Genome maintenance mechanisms for preventing cancer. Nature

    411: 366-374.

    Hoffmann I, Draetta G and Karsenti E (1994) Activation of the phosphatase activity of

    human cdc25A by a cdk2-cyclin E dependent phosphorylation at the G1/S transition.

    EMBO J 13: 4302-4310.

    Hopker K, Hagmann H, Khurshid S, Chen S, Hasskamp P, Seeger-Nukpezah T, Schilberg K,

    Heukamp L, Lamkemeyer T, Sos M, Thomas R, Lowery D and Roels F (2012)

    AATF/Che-1 acts as a phosphorylation-dependent molecular modulator to repress

    p53-driven apoptosis. EMBO J 31: 3961-3975.

    Huang L, Min J-N, Masters S, Mivechi NF and Moskophidis D (2007) Insights into function

    and regulation of small heat shock protein 25 (HSPB1) in a mouse model with

    targeted gene disruption. Genesis 45: 487-501.

    Huang X, Zhu X, Chen X, Zhou W, Xiao D, Degrado S, Aslanian R, Fossetta J, Lundell D,

    Tian F, Trivedi P and Palani A (2012) A three-step protocol for lead optimization:

    Quick identification of key conformational features and functional groups in the SAR

    studies of non-ATP competitive MK2 (MAPKAPK2) inhibitors. Bioorg Med Chem

    Lett 22: 65-70.

    Ischia J and So AI (2013) The role of heat shock proteins in bladder cancer. Nature Rev

    Urology 10: 386-395.

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    36

    Jazayeri A, Falck J, Lukas C, Bartek J, Smith GC, Lukas J and Jackson SP (2006) ATM- and

    cell cycle-dependent regulation of ATR in response to DNA double-strand breaks.

    Nat Cell Biol 8: 37-45.

    Kaptein A, Oubrie A, Zwart Ed, Hoogenboom N, Wit Jd, Kar Bvd, Hoek Mv, Vogel G,

    Kimpe Vd, Schultz-Fademrecht C, Borsboom J, Zeeland Mv, Versteegh J, Kazemier

    B, Roos Jd, Wijnands F, Dulos J, Jaeger M, Leandro-Garcia P and Barf T (2011)

    Discovery of selective and orally available spiro-3-piperidyl ATP-competitive MK2

    inhibitors. Bioorg Med Chem Lett 21: 3823-3827.

    Kastan MB and Bartek J (2004) Cell-cycle checkpoints and cancer. Nature 432: 316-323.

    Kishi H, Nakagawa K, Matsumoto M, Suga M, Ando M, Taya Y and Yamaizumi M (2001)

    Osmotic shock induces G1 arrest through p53 phosphorylation at Ser33 by activated

    p38MAPK without phosphorylation at Ser15 and Ser20. J Biol Chem 276: 39115-

    39122.

    Kobayashi M, Nishita M, Mishima T, Ohashi K and Mizuno K (2006) MAPKAPK-2-

    mediated LIM-kinase activation is critical for VEGF-induced actin remodelling and

    cell migration. EMBO J 25: 713-726.

    Kobayashi Y, Qi X and Chen G (2012) MK2 regulates Ras oncogenesis through stimulation

    ROS production. Genes Cancer 3: 521-530.

    Köpper F, Bierwirth C, Schön M, Kunze M, Elvers I, Kranz D, Saini P, Menon MB, Walter

    D, Sørensen CS, Gaestel M, Helleday T, Schön MP and Dobbelstein M (2013)

    Damage-induced DNA replication stalling relies on MAPK-activated protein kinase 2

    activity. Proc Natl Acad Sci USA 110: 16856-16861.

    Kostenko S and Moens U (2009) Heat shock protein 27 phosphorylation: kinases,

    phosphatases, functions and pathology. Cell Mol Life Sci 66: 3289-3307.

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    37

    Kotlyarov A, Neininger A, Schubert C, Eckert R, Birchmeier C, Volk HD and Gaestel M

    (1999) MAPKAP kinase 2 is essential for LPS-induced TNF-alpha biosynthesis. Nat

    Cell Biol 1: 94-97.

    Kotlyarov A, Yannoni Y, Fritz S, Laaß K, Telliez J-B, Pitman D, Lin L-L and Gaestel M

    (2002) Distinct Cellular Functions of MK2. Mol Cell Biol 22: 4827-4835.

    Kukkonen-Macchi A, Sicora O, Kaczynska K, Oetken-Lindholm C, Pouwels J, Laine L and

    Kallio MJ (2011) Loss of p38gamma MAPK induces pleiotropic mitotic defects and

    massive cell death. J Cell Sci 124: 216-227.

    Kumagai A, Kim SM and Dunphy WG (2004) Claspin and the activated form of ATR-

    ATRIP collaborate in the activation of Chk1. J Biol Chem 279: 49599-49608.

    Kumar B, Koul S, Petersen J, Khandrika L, Hwa JS, Meacham RB, Wilson S and Koul HK

    (2010) p38 Mitogen-activated protein kinase–driven MAPKAPK2 regulates invasion

    of bladder cancer by modulation of MMP-2 and MMP-9 activity. Cancer Res 70: 832-

    841.

    Lafarga V, Cuadrado A, Lopez de Silanes I, Bengoechea R, Fernandez-Capetillo O and

    Nebreda AR (2009) p38 Mitogen-activated protein kinase- and HuR-dependent

    stabilization of p21(Cip1) mRNA mediates the G(1)/S checkpoint. Mol Cell Biol 29:

    4341-4351.

    Lam MH, Liu Q, Elledge SJ and Rosen JM (2004) Chk1 is haploinsufficient for multiple

    functions critical to tumor suppression. Cancer Cell 6: 45-59.

    Lammer C, Wagerer S, Saffrich R, Mertens D, Ansorge W and Hoffmann I (1998) The

    cdc25B phosphatase is essential for the G2/M phase transition in human cells. J Cell

    Sci 111: 2445-2453.

    This article has not been copyedited and formatted. The final version may differ from this version.Molecular Pharmacology Fast Forward. Published on December 2, 2013 as DOI: 10.1124/mol.113.090365

    at ASPE

    T Journals on July 5, 2021

    molpharm

    .aspetjournals.orgD

    ownloaded from

    http://molpharm.aspetjournals.org/

  • MOL #90365

    38

    Lasa M, Mahtani KR, Finch A, Brewer G, Saklatvala J and Clark AR (2000) Regulation of

    cyclooxygenase 2 mRNA stability by the mitogen-activated protein kinase p38

    signaling cascade. Mol Cell Biol 20: 4265-4274.

    Lemaire M, Froment C, Boutros R, Mondesert O, Nebreda AR, Monsarrat B and Ducommun

    B (2006) CDC25B phosphorylation by p38 and MK-2. Cell Cycle 5: 1649-1653.

    Liu Q, Guntuku S, Cui XS, Matsuoka S, Cortez D, Tamai K, Luo G, Carattini-Rivera S,

    DeMayo F, Bradley A, Donehower LA and Elledge SJ (2000) Chk1 is an essential

    kinase that is regulated by Atr and required for the G(2)/M DNA damage checkpoint.

    Genes Dev 14: 1448-1459.

    Liu X and Erikson RL (2003) Polo-like kinase (Plk)1 depletion induces apoptosis in cancer

    cells. Proc Natl Acad Sci U S A 100: 5789-5794.

    Llopis A, Salvador N, Ercilla A, Guaita-Esteruelas S, Barrantes Idel B, Gupta J, Gaestel M,

    Davis RJ, Nebreda AR and Agell N (2012) The stress-activated protein kinases

    p38alpha/beta and JNK1/2 cooperate with Chk1 to inhibit mitotic entry upon DNA

    replication arrest. Cell Cycle 11: 3627-3637.

    Manke IA, Nguyen A, Lim D, Stewart MQ, Elia AE and Yaffe MB (2005) MAPKAP kinase-

    2 is a cell cycle checkpoint kinase that regulates the G2/M transition and S phase

    progression in response to UV irradiation. Mol Cell 17: 37-48.

    Marotta D, Karar J, Jenkins WT, Kumanova M, Jenkins KW, Tobias JW, Baldwin D,

    Hatzigeorgiou A, Alexiou P, Evans SM, Alarcon R, Maity A, Koch C and Koumenis

    C (2011) In vivo profiling of hypoxic gene expression in gliomas using the hypoxia

    marker EF5 and laser-capture microdissection. Cancer Res 71: 779-789.

    Matsuoka S, Huang M and Elledge SJ (1998) Linkage of ATM to cell cycle regulation by the

    Chk2 protein kinase. Science 282: 1893-1897.

    This article has no