The ATM kinase inhibitor KU-55933 provides neuroprotection against hydrogen peroxide-induced cell damage via a γH2AX/p-p53/caspase-3-independent mechanism: Inhibition of calpain and cathepsin D

https://doi.org/10.1016/j.biocel.2017.03.015Get rights and content

Highlights

  • KU-55933 provided neuroprotection against H2O2- and doxorubicin (Dox)- but not staurosporine (St)-induced cell damage.

  • KU-55933 mediated greater protection in neuronallly differentiated SH-SY5Y cells.

  • KU-55933 blocked phosphorylation of ATM in H2O2 and Dox models of cell damage.

  • KU-55933 prevented the toxin induced increases in γH2AX, p-p53 and caspase-3 only in the Dox model.

  • KU-55933 attenuated the H2O2-induced increases in calpain and cathepsin D activity.

Abstract

The role of the kinase ataxia-telangiectasia mutated (ATM), a well-known protein engaged in DNA damage repair, in the regulation of neuronal responses to oxidative stress remains unexplored. Thus, the neuroprotective efficacy of KU-55933, a potent inhibitor of ATM, against cell damage evoked by oxidative stress (hydrogen peroxide, H2O2) has been studied in human neuroblastoma SH-SY5Y cells and compared with the efficacy of this agent in models of doxorubicin (Dox)- and staurosporine (St)-evoked cell death. KU-55933 inhibited the cell death induced by H2O2 or Dox but not by St in undifferentiated (UN-) and retinoic acid-differentiated (RA)-SH-SY5Y cells, with a more pronounced effect in the latter cell phenotype. Furthermore, this ATM inhibitor attenuated the Dox- but not H2O2-induced caspase-3 activity in both UN- and RA-SH-SY5Y cells. Although KU-55933 inhibited the H2O2- and Dox-induced activation of ATM, it attenuated the toxin-induced phosphorylation of the proteins H2AX and p53 only in the latter model of cell damage. Moreover, the ATM inhibitor prevented the H2O2-evoked increases in calpain and cathepsin D activity and attenuated cell damage to a similar degree as inhibitors of calpain (MDL28170) and cathepsin D (pepstatin A). Finally, we confirmed the neuroprotective potential of KU-55933 against the H2O2- and Dox-evoked cell damage in primary mouse cerebellar granule cells and in the mouse hippocampal HT-22 cell line. Altogether, our results extend the neuroprotective portfolio of KU-55933 to a model of oxidative stress, with this effect not involving inhibition of the γH2AX/p-p53/caspase-3 pathway and instead associated with the attenuation of calpain and cathepsin D activity.

Introduction

The ataxia telangiectasia mutated (ATM, EC 2.7.11.1) kinase, as a member of the family of PI3-Ks (phosphatidylinositol 3-kinases), regulates a wide variety of processes, including control of gene expression and the cell cycle, chromatin organization, stress response, cell metabolism and intracellular organization (Shiloh and Ziv, 2013). Loss-of-function mutation in ATM is a cause of ataxia telangiectasia syndrome (A-T) manifested inter alia by impaired coordination, increased risk of cancer, enlarged blood vessels in the skin and eyeballs, immunodeficiency and neurodegeneration (Lee and McKinnon, 2007, McKinnon, 2012). The first identified role of ATM was participation in the response to double strand breakage (DSB), which is thought to be the most detrimental form of DNA damage in mammalian cells (Valerie and Povirk, 2003). When cell homeostasis is preserved, ATM exists in the form of an inactive dimer/tetramer, but in the presence of DSB, ATM undergoes autophosphorylation at Ser1981 (pATM), forms a monomer/dimer and migrates to the nucleus. Histone H2AX is one of the first ATM substrates, and H2AX phosphorylated at Ser139 (γH2AX) is a molecular marker of DSB. Through NBS1 (Nijmegen breakage syndrome 1) protein, ATM kinase is linked with the MRN (MRE11-RAD50-NBS1) complex at the sites of DNA strand breaks, where it phosphorylates proteins, such as the serine/threonine-protein kinase Chk2 (at Thr68) and p53 (at Ser15). This leads to cell cycle arrest and repair of damage, or to apoptosis if the damage is too severe (Bakkenist and Kastan, 2003, Martin, 2008, Stracker et al., 2013). The mechanisms and mutual interconnections between DNA damage and repair, cell cycle regulation, and induction of apoptosis in proliferating cells are quite well known (Iijima et al., 2008a, Iijima et al., 2008b, Matt and Hofmann, 2016, Shiloh and Ziv, 2013); however, these processes are less clear in post-mitotic neurons (Kruman et al., 2004, Lee and McKinnon, 2007, Martin and Wong, 2016). Nevertheless, there are clinical and experimental data demonstrating that the DNA damage-evoked cell-cycle re-entry and apoptosis are important factors contributing to the neuronal cell loss observed in various neurodegenerative disorders (e.g., Alzheimer’s disease, Parkinson’s disease, Huntington's disease and amyotrophic lateral sclerosis) (Camins et al., 2010, Martin, 2008, Mullaart et al., 1990, Pelegrí et al., 2008, Ranganathan and Bowser, 2003, Silva et al., 2014, Smith et al., 2004). Moreover, it has been shown that not only various genotoxic stimuli (e.g., γ-irradiation, camptothecin, etoposide, doxorubicin, methotrexate, homocysteine) but also factors involved in pathomechanisms of neurodegeneration (e.g., β-amyloid, N-methyl-d-aspartate) and some neurotoxins (e.g., 6-hydroxydopamine (6-OHDA), 1-methyl-4-phenylpyridinium iodide (MPP(+)), N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4)) could induce neuronal cell death via DNA damage-induced cell-cycle re-entry and/or apoptosis (Alvira et al., 2007, Bernstein et al., 2011, Camins et al., 2010, Jung et al., 2011; Macleod et al., 2003, Wang et al., 2014, Wang et al., 2015a). Additionally, there are data linking neuroprotective effects of some drugs with the inhibition of DNA damage, as shown for some antidepressant drugs in the DSP4- or camptothecin-induced models of SH-SY5Y cell damage (Wang et al., 2015b).

Although various experimental studies have demonstrated neuroprotective effects of the inhibition of cell-cycle re-entry (e.g., by cyclin-dependent kinase 5 (CDK5) inhibitors) (Cicenas et al., 2015, Neve and McPhie, 2006, Pizarro et al., 2011), the role of the functional modulation of ATM kinase in neuroprotection remains less recognized. Until 2004, when the selective ATM kinase inhibitor KU-55933 was designed (Hickson et al., 2004), non-specific inhibitors such as caffeine or wortmannin were used to inhibit the activity of ATM (Adams et al., 2010, Burma et al., 2001, Guo et al., 2010, Kruman et al., 2004). Among the limited studies on the neuroprotection mediated by ATM inhibition, there are data demonstrating beneficial effects of KU-55933 and/or caffeine in the models of neuronal cell damage induced by MPP(+), β-amyloid or DNA-damaging drugs (etoposide and homocysteine) with mechanisms involving DSB (γH2AX, p-p53), apoptosis and/or cell cycle regulatory proteins (Alvira et al., 2007, Camins et al., 2010, Jung et al., 2011). On the other hand, there are reports suggesting a protective role for ATM based on findings from A-T fibroblasts, which were more sensitive to hydrogen peroxide (H2O2)-induced apoptosis (Yu et al., 2015), as well as the observations from Atm-deficient mice of increased oxidative damage in brain tissues (particularly cerebellar Purkinje cells) and loss of dopamine neurons in the nigrostriatal pathway (Barlow et al., 1999, Kirshner et al., 2012). According to current knowledge, the oxidation-evoked ATM activation appear to involve different mechanisms than this one induced by DSB. For example, human fibroblasts treated with H2O2 formed active, linked by disulfide bonds ATM dimers which did not phosphorylate H2AX (Guo et al., 2010).

Since the elevated level of reactive oxygen species (ROS) is considered an important factor contributing to neuronal cell loss under various neurodegenerative conditions (Bredesen et al., 2006, Trippier et al., 2013), an investigation of the still-unknown role of ATM kinase in the neuronal response to oxidative stress is of high scientific and clinical importance. Thus, in the present study, we tested the effect of the specific inhibitor of ATM kinase, KU-55933, against H2O2-induced neuronal cell damage. Moreover, we compared the obtained results with the efficacy of this agent in the models of neurotoxicity induced by doxorubicin (Dox), a DNA damaging agent and/or inducer of the extracellular apoptotic pathway (Jantas et al., 2008, Jantas and Lason, 2009a, Lopes et al., 2008), and staurosporine (St), an inducer of the mitochondrial apoptotic pathway (Jantas et al., 2009, Koh et al., 1995). Our study, particularly the mechanistic components, was performed in human neuroblastoma SH-SY5Y cells, a commonly used cell line in neurotoxicity/neuroprotection research (Cheung et al., 2009; Lopes et al., 2010, Presgraves et al., 2004). Since there is still ongoing discussion regarding whether SH-SY5Y cells should be differentiated into neurons for neuroprotection studies (Cheung et al., 2009, Jantas et al., 2013, Lopes et al., 2010, Luchtman and Song, 2010, Presgraves et al., 2004, Wenker et al., 2010, Yong-Kee et al., 2011), we used undifferentiated (UN-) and retinoic acid-differentiated (RA)-SH-SY5Y cells. Finally, we confirmed data obtained in human SH-SY5Y cells in mouse primary cerebellar granule cells (CGCs) and the cell lines HT-22 (immortalized mouse hippocampal cells) and C6 (rat glioma).

Section snippets

Chemicals

Dulbecco’s modified Eagle medium (DMEM), FluoroBrite™ DMEM, fetal bovine serum (FBS), Neurobasal A medium, and supplement B27 (without antioxidants) were purchased from Gibco (Invitrogen, Poisley, UK). The Cytotoxicity Detection Kit (LDH) and BM Chemiluminescence Western Blotting Kit were purchased from Roche Diagnostics (Mannheim, Germany). KU-55933 (2-(4-Morpholinyl)-6-(1-thianthrenyl)-4H-pyran-4-one) and NU-7441 were purchased from Adooq Bioscience (Irvine, CA, USA) and Tocris (Bristol, UK),

KU-55933 at higher concentrations is toxic for SH-SY5Y cells

Treating UN- or RA-SH-SY5Y cells with lower concentrations (up to 1 μM) of KU-55933 for 24 h did not evoke any harmful effects, as confirmed by biochemical LDH release and MTT reduction assays as well as by flow cytometric analysis of PI-positive nuclei (Table 1). At 10 μM, the ATM inhibitor significantly increased LDH release in UN- and RA-SH-SY5Y cells but had no effect on the MTT reduction and PI assays (Table 1). However, the highest concentration of KU-55933 tested, 20 μM, decreased the

Discussion

To our knowledge, this is the first study demonstrating neuroprotective effects of the ATM inhibitor KU-55933 against cell damage induced by the oxidative stress inducer H2O2. We obtained these results in various neuronal cell cultures (mouse primary cerebellar granule cells (CGCs), human neuroblastoma SH-SY5Y cells and HT-22 immortalized mouse hippocampal cells) but not in C6 rat glioma cells, suggesting that the protective effects of the ATM inhibitor against oxidative stress-induced cell

Conflict of interest

The authors declare that they have no conflicts of interest with this work.

Author contributions

JC, DJ and WL designed experiments; JC and DJ carried out experiments; JC analyzed experimental results; DJ and JC wrote the manuscript; WL critically reviewed the first draft of manuscript and suggested improvements.

Acknowledgments

The study was supported by statutory funds of the Institute of Pharmacology, Polish Academy of Sciences. Jakub Chwastek holds a scholarship from the KNOW, sponsored by the Ministry of Science and Higher Education, Republic of Poland. We kindly thank Ms. Barbara Korzeniak for her excellent technical assistance.

References (108)

  • D. Jantas et al.

    The attenuating effect of memantine on staurosporine-, salsolinol- and doxorubicin-induced apoptosis in human neuroblastoma SH-SY5Y cells

    Neurochem. Int.

    (2008)
  • D. Jantas et al.

    The neuroprotective effects of orthosteric agonists of group II and III mGluRs in primary neuronal cell cultures are dependent on developmental stage

    Neuropharmacology

    (2016)
  • L. Jaworska-Feil et al.

    Protective effects of TRH and its analogues against various cytotoxic agents in retinoic acid (RA)-differentiated human neuroblastoma SH-SY5Y cells

    Neuropeptides

    (2010)
  • M. Katsuyama et al.

    Clioquinol induces DNA double-strand breaks, activation of ATM, and subsequent activation of p53 signaling

    Toxicology

    (2012)
  • I.I. Kruman et al.

    Cell cycle activation linked to neuronal cell death initiated by DNA damage

    Neuron

    (2004)
  • X. Kuang et al.

    Activation of AMP-activated protein kinase in cerebella of Atm-/- mice is attributable to accumulation of reactive oxygen species

    Biochem. Biophys. Res. Commun.

    (2012)
  • Y. Lee et al.

    Responding to DNA double strand breaks in the nervous system

    Neuroscience

    (2007)
  • E. Lee et al.

    Effect of conjugated linoleic acid, μ-calpain inhibitor, on pathogenesis of Alzheimer’s disease

    Biochim. Biophys. Acta

    (2013)
  • C.T. Lee et al.

    Nitroxide antioxidant as a potential strategy to attenuate the oxidative/nitrosative stress induced by hydrogen peroxide plus nitric oxide in cultured neurons

    Nitric Oxide

    (2016)
  • C.S. Lin et al.

    Autophagy and reactive oxygen species modulate cytotoxicity induced by suppression of ATM kinase activity in head and neck cancer cells

    Oral Oncol.

    (2012)
  • M.A. Lopes et al.

    Doxorubicin induces biphasic neurotoxicity to rat cortical neurons

    Neurotoxicology

    (2008)
  • F.M. Lopes et al.

    Comparison between proliferative and neuron-like SH-SY5Y cells as an in vitro model for Parkinson disease studies

    Brain Res.

    (2010)
  • D.W. Luchtman et al.

    Why SH-SY5Y cells should be differentiated

    Neurotoxicology

    (2010)
  • E. Mullaart et al.

    Increased levels of DNA breaks in cerebral cortex of Alzheimer's disease patients

    Neurobiol. Aging

    (1990)
  • R.L. Neve et al.

    The cell cycle as a therapeutic target for Alzheimer's disease

    Pharmacol. Ther.

    (2006)
  • T. Pan et al.

    Rapamycin protects against rotenone-induced apoptosis through autophagy induction

    Neuroscience

    (2009)
  • C. Pelegrí et al.

    Cell cycle activation in striatal neurons from Huntington’s disease patients and rats treated with 3-nitropropionic acid

    Int. J. Dev. Neurosci.

    (2008)
  • N. Raimundo et al.

    Mechanisms of communication between mitochondria and lysosomes

    Int. J. Biochem. Cell Biol.

    (2016)
  • S. Ranganathan et al.

    Alterations in G(1) to S phase cell-cycle regulators during amyotrophic lateral sclerosis

    Am. J. Pathol.

    (2003)
  • M. Richter et al.

    SK channel activation modulates mitochondrial respiration and attenuates neuronal HT-22 cell damage induced by H2O2

    Neurochem. Int.

    (2015)
  • C.R. Rocourt et al.

    The catalytic subunit of DNA-dependent protein kinase is downstream of ATM and feeds forward oxidative stress in the selenium-induced senescence response

    J. Nutr. Biochem.

    (2013)
  • S. Sahara et al.

    Calpain-mediated Hsp70.1 cleavage in hippocampal CA1 neuronal death

    Biochem. Biophys. Res. Commun.

    (2010)
  • P.D. Smith et al.

    CDKs: taking on a role as mediators of dopaminergic loss in Parkinson’s disease

    Trends Mol. Med.

    (2004)
  • A.S. Unnithan et al.

    N-acetyl cysteine prevents synergistic, severe toxicity from two hits of oxidative stress

    Neurosci. Lett.

    (2014)
  • I.M. Ward et al.

    Histone H2AX Is Phosphorylated in an ATR dependent Manner in Response to Replicational Stress

    J. Biol. Chem.

    (2001)
  • B.R. Adams et al.

    Dynamic dependence on ATR and ATM for double-strand break repair in human embryonic stem cells and neural descendants

    PLoS One

    (2010)
  • D. Alvira et al.

    Neuroprotective effects of caffeine against complex I inhibition-induced apoptosis are mediated by inhibition of the Atm/p53/E2F-1 path in cerebellar granule neurons

    J. Neurosci. Res.

    (2007)
  • C.J. Bakkenist et al.

    DNA damage activates ATM through intermolecular autophosphorylation and dimer dissociation

    Nature

    (2003)
  • C. Barlow et al.

    Loss of the ataxia-telangiectasia gene product causes oxidative damage in target organs

    Proc. Natl. Acad. Sci. U. S. A.

    (1999)
  • C. Barlow et al.

    ATM is a cytoplasmic protein in mouse brain required to prevent lysosomal accumulation

    Proc. Natl. Acad. Sci. U. S. A.

    (2000)
  • E. Berkovich et al.

    Roles of ATM and NBS1 in chromatin structure modulation and DNA double-strand break repair

    Nat. Cell. Biol.

    (2007)
  • A.I. Bernstein et al.

    6-OHDA generated ROS induces DNA damage and p53- and PUMA-dependent cell death

    Mol. Neurodegener.

    (2011)
  • J.K. Boehrs et al.

    Constitutive expression and cytoplasmic compartmentalization of ATM protein in differentiated human neuron-like SH-SY5Y cells

    J. Neurochem.

    (2007)
  • D.E. Bredesen et al.

    Cell death in the nervous system

    Nature

    (2006)
  • A. Camins et al.

    Activation of ataxia telangiectasia muted under experimental models and human Parkinson’s disease

    Cell. Mol. Life. Sci.

    (2010)
  • J. Cicenas et al.

    Roscovitine in cancer and other diseases

    Ann. Transl. Med.

    (2015)
  • N. Doti et al.

    Inhibition of the AIF/CypA complex protects against intrinsic death pathways induced by oxidative stress

    Cell. Death Dis.

    (2014)
  • C.R. Foster et al.

    Deficiency of ataxia telangiectasia mutated kinase modulates cardiac remodeling following myocardial infarction: involvement in fibrosis and apoptosis

    PLoS One

    (2013)
  • S.E. Golding et al.

    Extracellular signal-related kinase positively regulates ataxia telangiectasia mutated, homologous recombination repair, and the DNA damage response

    Cancer Res.

    (2007)
  • S.E. Golding et al.

    Improved ATM kinase inhibitor KU-60019 radiosensitizes glioma cells, compromises insulin, AKT and ERK prosurvival signaling, and inhibits migration and invasion

    Mol. Cancer Ther.

    (2009)
  • Cited by (0)

    View full text