A chemical with proven clinical safety rescues Down ...A chemical with proven clinical safety...

10
RESEARCH ARTICLE A chemical with proven clinical safety rescues Down-syndrome- related phenotypes in through DYRK1A inhibition Hyeongki Kim 1,2, *, Kyu-Sun Lee 3,4, *, Ae-Kyeong Kim 3 , Miri Choi 1 , Kwangman Choi 1 , Mingu Kang 1 , Seung-Wook Chi 5 , Min-Sung Lee 5 , Jeong-Soo Lee 3,4 , So-Young Lee 6 , Woo-Joo Song 7 , Kweon Yu 3,4 and Sungchan Cho 1,2, ABSTRACT DYRK1A is important in neuronal development and function, and its excessive activity is considered a significant pathogenic factor in Down syndrome and Alzheimers disease. Thus, inhibition of DYRK1A has been suggested to be a new strategy to modify the disease. Very few compounds, however, have been reported to act as inhibitors, and their potential clinical uses require further evaluation. Here, we newly identify CX-4945, the safety of which has been already proven in the clinical setting, as a potent inhibitor of DYRK1A that acts in an ATP-competitive manner. The inhibitory potency of CX-4945 on DYRK1A (IC 50 =6.8 nM) in vitro was higher than that of harmine, INDY or proINDY, which are well-known potent inhibitors of DYRK1A. CX-4945 effectively reverses the aberrant phosphorylation of Tau, amyloid precursor protein (APP) and presenilin 1 (PS1) in mammalian cells. To our surprise, feeding with CX-4945 significantly restored the neurological and phenotypic defects induced by the overexpression of minibrain, an ortholog of human DYRK1A, in the Drosophila model. Moreover, oral administration of CX-4945 acutely suppressed Tau hyperphosphorylation in the hippocampus of DYRK1A-overexpressing mice. Our research results demonstrate that CX-4945 is a potent DYRK1A inhibitor and also suggest that it has therapeutic potential for DYRK1A-associated diseases. KEY WORDS: Down syndrome, Alzheimers disease, DYRK1A, CX-4945, Tau hyperphosphorylation INTRODUCTION Dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) is a serine/threonine kinase important to brain development. DYRK1A plays a variety of functional roles within the adult central nervous system (Tejedor and Hammerle, 2011). The significance of DYRK1A has been recently highlighted by the discovery of its contribution to Down syndrome (DS) pathogenesis. DS is the most common genetic disorder, with an incidence of 1 in 800 live births, and is caused by a complete or partial trisomy of chromosome 21. DS is characterized by various symptoms, including mental retardation and congenital heart defects, as well as by defects in immune and endocrine systems (Becker and Joost, 1999). These featured phenotypes are closely associated with the overexpression or hyperactivity of many genes that can be mapped within the Down syndrome critical region (DSCR) on chromosome 21 (Guimera et al., 1999; Wiseman et al., 2009). In particular, mental retardation, which is a characteristic symptom of DS, is thought to be related primarily to the DYRK1A gene in the DSCR (Smith and Rubin, 1997). Many studies using different lines of transgenic mice have shown that the additional expression of DYRK1A in a normal mouse, which mimics trisomy in human DS, is sufficient to cause abnormalities in learning and memory as well as brain structure, strongly suggesting a central function for DYRK1A in the mental retardation associated with DS (Ahn et al., 2006; Altafaj et al., 2001). Moreover, mice with lowered DYRK1A expression show phenotypic effects similar to those in mice overexpressing DYRK1A, indicating that DYRK1A activity is tightly controlled during normal brain development and that a dosage imbalance in DYRK1A expression affects brain structure and function (Arque et al., 2008; Benavides-Piccione et al., 2005; Fotaki et al., 2002, 2004). Intriguingly, increased DYRK1A activity has been also reported in various brain compartments in subjects that suffer from Alzheimers disease (AD), a representative neurodegenerative disease (Ferrer et al., 2005; Tiraboschi et al., 2004). At the neuropathological level, DS and AD share several features that are characterized by the presence of amyloid plaques and neurofibrillary tangles (NFTs), the formation of which is affected by the aberrant phosphorylation of Tau (for NFTs), as well as of amyloid precursor protein (APP) and presenilin 1 (PS1) (for amyloid plaques) (Johnson and Hartigan, 1999; Tiraboschi et al., 2004). Moreover, it has been reported that DYRK1A directly phosphorylates Tau, APP and PS1 (Ryoo et al., 2008, 2007; Ryu et al., 2010). These observations provide a plausible link between DS and AD that could explain the early onset of AD-like symptoms in the majority of people with DS and further indicate that DYRK1A could be a promising therapeutic target for treating diseases such as DS and AD that involve DYRK1A overexpression or hyperactivity. Despite substantial efforts to develop potent and selective inhibitors of DYRK1A, only a few are currently available, and their potential clinical use remains to be tested further (Smith et al., 2012). Extensive evaluations of the most promising DYRK1A Received 5 April 2016; Accepted 16 June 2016 1 Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungbuk 28115, Republic of Korea. 2 Department of Biomolecular Science, University of Science and Technology, Daejeon 34113, Republic of Korea. 3 Neurophysiology Research Group, Hazard Monitoring BioNano Research Center, Korea Research Institute of Bioscience and Biotechnology, Deajeon 34141, Republic of Korea. 4 Department of Functional Genomics, University of Science and Technology, Daejeon 34113, Republic of Korea. 5 Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea. 6 International Cooperation Office, Ministry of Food & Drug Safety, Cheongju, Chungbuk 28159, Republic of Korea. 7 Department of Biochemistry and Molecular Biology, Neurodegeneration Control Research Center, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea. *These authors contributed equally to this work Author for correspondence ([email protected]) S.C., 0000-0001-7627-978X This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed. 839 © 2016. Published by The Company of Biologists Ltd | Disease Models & Mechanisms (2016) 9, 839-848 doi:10.1242/dmm.025668 Disease Models & Mechanisms

Transcript of A chemical with proven clinical safety rescues Down ...A chemical with proven clinical safety...

Page 1: A chemical with proven clinical safety rescues Down ...A chemical with proven clinical safety rescues Down-syndrome-related phenotypes in through DYRK1A inhibition Hyeongki Kim1,2,*,

RESEARCH ARTICLE

A chemical with proven clinical safety rescues Down-syndrome-related phenotypes in through DYRK1A inhibitionHyeongki Kim1,2,*, Kyu-Sun Lee3,4,*, Ae-Kyeong Kim3, Miri Choi1, Kwangman Choi1, Mingu Kang1,Seung-Wook Chi5, Min-Sung Lee5, Jeong-Soo Lee3,4, So-Young Lee6, Woo-Joo Song7, Kweon Yu3,4

and Sungchan Cho1,2,‡

ABSTRACTDYRK1A is important in neuronal development and function, and itsexcessive activity is considered a significant pathogenic factor in Downsyndrome and Alzheimer’s disease. Thus, inhibition of DYRK1A hasbeen suggested to be a new strategy to modify the disease. Very fewcompounds, however, have been reported to act as inhibitors, and theirpotential clinical uses require further evaluation. Here, we newly identifyCX-4945, the safety of which has been already proven in the clinicalsetting, as a potent inhibitor of DYRK1A that acts in an ATP-competitivemanner. The inhibitory potency of CX-4945 on DYRK1A (IC50=6.8 nM)in vitro was higher than that of harmine, INDY or proINDY, which arewell-known potent inhibitors of DYRK1A. CX-4945 effectively reversesthe aberrant phosphorylation of Tau, amyloid precursor protein (APP)andpresenilin 1 (PS1) inmammalian cells. To our surprise, feedingwithCX-4945 significantly restored the neurological and phenotypic defectsinduced by the overexpression of minibrain, an ortholog of humanDYRK1A, in the Drosophila model. Moreover, oral administration ofCX-4945 acutely suppressed Tau hyperphosphorylation in thehippocampus of DYRK1A-overexpressing mice. Our research resultsdemonstrate that CX-4945 is a potent DYRK1A inhibitor and alsosuggest that it has therapeutic potential for DYRK1A-associateddiseases.

KEY WORDS: Down syndrome, Alzheimer’s disease, DYRK1A,CX-4945, Tau hyperphosphorylation

INTRODUCTIONDual-specificity tyrosine phosphorylation-regulated kinase 1A(DYRK1A) is a serine/threonine kinase important to brain

development. DYRK1A plays a variety of functional roles withinthe adult central nervous system (Tejedor and Hammerle, 2011).The significance of DYRK1A has been recently highlighted by thediscovery of its contribution to Down syndrome (DS) pathogenesis.

DS is the most common genetic disorder, with an incidence of1 in 800 live births, and is caused by a complete or partial trisomyof chromosome 21. DS is characterized by various symptoms,including mental retardation and congenital heart defects, as well asby defects in immune and endocrine systems (Becker and Joost,1999). These featured phenotypes are closely associated with theoverexpression or hyperactivity of many genes that can be mappedwithin the Down syndrome critical region (DSCR) on chromosome21 (Guimera et al., 1999; Wiseman et al., 2009). In particular,mental retardation, which is a characteristic symptom of DS, isthought to be related primarily to the DYRK1A gene in the DSCR(Smith and Rubin, 1997). Many studies using different lines oftransgenic mice have shown that the additional expression ofDYRK1A in a normal mouse, which mimics trisomy in human DS,is sufficient to cause abnormalities in learning and memory as wellas brain structure, strongly suggesting a central function forDYRK1A in the mental retardation associated with DS (Ahnet al., 2006; Altafaj et al., 2001). Moreover, mice with loweredDYRK1A expression show phenotypic effects similar to those inmice overexpressing DYRK1A, indicating that DYRK1A activity istightly controlled during normal brain development and that adosage imbalance in DYRK1A expression affects brain structureand function (Arque et al., 2008; Benavides-Piccione et al., 2005;Fotaki et al., 2002, 2004).

Intriguingly, increased DYRK1A activity has been also reportedin various brain compartments in subjects that suffer fromAlzheimer’s disease (AD), a representative neurodegenerativedisease (Ferrer et al., 2005; Tiraboschi et al., 2004). At theneuropathological level, DS and AD share several features that arecharacterized by the presence of amyloid plaques andneurofibrillary tangles (NFTs), the formation of which is affectedby the aberrant phosphorylation of Tau (for NFTs), as well as ofamyloid precursor protein (APP) and presenilin 1 (PS1) (foramyloid plaques) (Johnson and Hartigan, 1999; Tiraboschi et al.,2004). Moreover, it has been reported that DYRK1A directlyphosphorylates Tau, APP and PS1 (Ryoo et al., 2008, 2007; Ryuet al., 2010). These observations provide a plausible link betweenDS and AD that could explain the early onset of AD-like symptomsin the majority of peoplewith DS and further indicate that DYRK1Acould be a promising therapeutic target for treating diseases such asDS and AD that involve DYRK1A overexpression or hyperactivity.

Despite substantial efforts to develop potent and selectiveinhibitors of DYRK1A, only a few are currently available, andtheir potential clinical use remains to be tested further (Smith et al.,2012). Extensive evaluations of the most promising DYRK1AReceived 5 April 2016; Accepted 16 June 2016

1Anticancer Agent Research Center, Korea Research Institute of Bioscience andBiotechnology, Cheongju, Chungbuk 28115, Republic of Korea. 2Department ofBiomolecular Science, University of Science and Technology, Daejeon 34113,Republic of Korea. 3Neurophysiology Research Group, Hazard MonitoringBioNano Research Center, Korea Research Institute of Bioscience andBiotechnology, Deajeon 34141, Republic of Korea. 4Department of FunctionalGenomics, University of Science and Technology, Daejeon 34113, Republic ofKorea. 5Disease Target Structure Research Center, Korea Research Institute ofBioscience and Biotechnology, Daejeon 34141, Republic of Korea. 6InternationalCooperation Office, Ministry of Food & Drug Safety, Cheongju, Chungbuk 28159,Republic of Korea. 7Department of Biochemistry and Molecular Biology,Neurodegeneration Control Research Center, School of Medicine, Kyung HeeUniversity, Seoul 02447, Republic of Korea.*These authors contributed equally to this work

‡Author for correspondence ([email protected])

S.C., 0000-0001-7627-978X

This is an Open Access article distributed under the terms of the Creative Commons AttributionLicense (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use,distribution and reproduction in any medium provided that the original work is properly attributed.

839

© 2016. Published by The Company of Biologists Ltd | Disease Models & Mechanisms (2016) 9, 839-848 doi:10.1242/dmm.025668

Disea

seModels&Mechan

isms

Page 2: A chemical with proven clinical safety rescues Down ...A chemical with proven clinical safety rescues Down-syndrome-related phenotypes in through DYRK1A inhibition Hyeongki Kim1,2,*,

inhibitors that have been developed to date suggest that theirtherapeutic application might still be limited by pharmacologicalside effects.Here, we report CX-4945 as a novel inhibitor of DYRK1Awith a

high potency. Its strong inhibitory effect on DYRK1A has beenextensively confirmed in vitro, in mammalian cells and even inliving organisms. Moreover, we pharmacologically validated theuse of CX-4945 in vivo in model organisms by observing theeffective rescue of neurological and phenotypic defects in a DS-likeDrosophila model, and the significant suppression of Tauphosphorylation in the hippocampus of DS-like mice. As a potentinhibitor of DYRK1Awith proven safety in clinical trials, CX-4945will be a valuable tool in DYRK1A-related basic research and in thedevelopment of therapeutic drugs for DYRK1A-associateddiseases, such as DS and AD.

RESULTSIdentification of CX-4945 as a novel inhibitor of DYRK1AOur recent research has demonstrated that CX-4945, a previouslywell-characterized inhibitor of casein kinase 2 (CK2) and amolecule currently in phase 1b and phase 2 clinical trials forcancer treatment, is a potent inhibitor (IC50=∼3-10 nM) of Cdc2-like kinases (Clks), which regulate alternative splicing (Kim et al.,2014; Siddiqui-Jain et al., 2010) (Fig. 1A). Intriguingly, manysmall-molecule inhibitors of Clks (TG-003, KH-CB19 andLeucettine L41) inhibit DYRKs with potencies similar to thosefor their inhibition of Clks (Debdab et al., 2011; Fedorov et al.,2011; Mott et al., 2009). This could be explained by thephylogenetic similarity between DYRKs and Clks (Aranda et al.,

2011; Kannan and Neuwald, 2004). In fact, along with CK2 andClks, DYRKs are classified as part of the CMGC superfamily ofproline- or arginine-directed serine/threonine kinases. Therefore, wetested whether CX-4945 also has an inhibitory effect on DYRKsusing in vitro kinase assays with human recombinant kinases and asynthetic peptide substrate (see In vitro kinase assays in Materialsand Methods). We found that CX-4945 potently inhibited theactivity of all DYRK-family proteins (IC50=6.8, 6.4, 18 and1500 nM for DYRK1A, DYRK1B, DYRK3 and DYRK4,respectively; Fig. 1B). Among them DYRK1A and DYRK1Bwere most strongly affected by CX-4945, and its potency was muchhigher (about 20-fold) than that of harmine, a potent DYRKinhibitor that is widely used (Adayev et al., 2011) (Fig. 1C). Amongthe DYRK-family proteins, DYRK1A is a considerable pathologicalfactor for DS; therefore, further studies were focused on theDYRK1A protein.

CX-4945 inhibits DYRK1A in an ATP-competitive mannerPreviously, CX-4945 has been found to inhibit the activities of CK2and Clks through binding to the ATP-binding pocket and competingwith ATP (Ferguson et al., 2011; Kim et al., 2014). Therefore, toexamine whether CX-4945 also acts in this manner, we measuredDYRK1A kinase activity with various combinations of ATP andCX-4945 concentrations in vitro. As a result, CX-4945 inhibitedDYRK1A in an ATP-competitive manner, as expected (Fig. 2A).Moreover, molecular docking studies using the CDOCKERprogram in Discovery Studio software were performed to gaindeeper insights into the orientation of CX-4945 in the ATP-bindingpocket of DYRK1A, and also into the interaction betweenDYRK1A and CX-4945 at the molecular level. Consequently, wefound that CX-4945 fits snugly into the ATP-binding pocket ofDYRK1A (Fig. 2B) and makes extensive hydrophobic interactionswith residues V173, A186, F238, M240, L241, L294 and V306 inDYRK1A. Additionally, four hydrogen bonds were predicted to bemade between CX-4945 and the adjacent residues (K188, L241 andD307) (Fig. 2C). In particular, the hydrogen bonds between thecarboxyl group of CX-4945 and the side-chain nitrogen of K188 inDYRK1A appear to be crucial for this binding, given that a similarhydrogen bond has also been observed for the binding of CX-4945to human protein kinase CK2α (Ferguson et al., 2011) (Fig. 2C).Overall, these results strongly support the notion that CX-4945directly inhibits DYRK1A through ATP competition.

CX-4945 is a potent inhibitor of DYRK1ANext, we examined whether CX-4945 also inhibits DYRK1A inmammalian cells. The microtubule-associated protein Tau is a well-characterized substrate of DYRK1A, and aberrant phosphorylationof this protein is associated with the formation of NFTs in DS andAD (Tiraboschi et al., 2004). Therefore, we first investigatedthe effect of CX-4945 on Tau phosphorylation. In particular,phosphorylation of Tau at T212, which has been prominentlyobserved in the brains of DYRK1A-overexpressing transgenic mice(Ryoo et al., 2007), was examined. Phosphorylation of Tau at T212was induced by the overexpression of DYRK1A in 293T cells andthen detected by western blotting with an antibody specific forphosphorylated Tau at Thr212. Under the same conditions, Tauphosphorylation was markedly decreased by treatment with CX-4945 in a dose-dependent manner with an IC50 of 100-200 nM(Fig. 3A and B). Moderate inhibition of Tau phosphorylation wasseen at 0.1 μM, and almost complete inhibition was achieved at1 μM. Importantly, the inhibitory efficacy of CX-4945 on DYRK1Awas significantly stronger than that of harmine, INDY and proINDY

Fig. 1. CX-4945 is a potent inhibitor of DYRK1A in vitro. (A) Diagram of thechemical structure of CX-4945. (B) Potent inhibition of DYRKs byCX-4945 wasobserved in in vitro kinase assays conducted by Life Technologies usingrecombinant human DYRK1A, DYRK1B, DYRK3 and DYRK4 proteins. Themean values and standard deviation (s.d.; error bars) were determined fromtwo independent assays. The IC50 values were determined using PRISMsoftware. (C) Inhibition of DYRKs activity by harmine was measured asdescribed for CX-4945 in B.

840

RESEARCH ARTICLE Disease Models & Mechanisms (2016) 9, 839-848 doi:10.1242/dmm.025668

Disea

seModels&Mechan

isms

Page 3: A chemical with proven clinical safety rescues Down ...A chemical with proven clinical safety rescues Down-syndrome-related phenotypes in through DYRK1A inhibition Hyeongki Kim1,2,*,

(IC50 values of ∼500, 2000 and 2000 nM, respectively), which arewell-known potent DYRK1A inhibitors (Ogawa et al., 2010; Smithet al., 2012) (Fig. S1). Similarly, CX-4945 strongly inhibited thephosphorylation of APP and PS1, which are also well-knownsubstrates of DYRK1A and are crucial for amyloid plaque formationin DS and AD (Ryu et al., 2010; Smith et al., 2012), with estimatedIC50 values of ∼80 and 100 nM for APP and PS1, respectively(Fig. 4). Collectively, these results clearly demonstrate that CX-4945is a potent DYRK1A inhibitor in vitro and in mammalian cells.

CX-4945 has a modulatory effect on DS- and AD-relatedcalcineurin-NFAT signalingBecause deregulation of calcineurin and nuclear factor of activatedT cells (NFAT) signaling is related to the development of a DS- andAD-like phenotype, and because DYRK1A plays an important rolein this signaling pathway (Arron et al., 2006), we examined theeffect of CX-4945 on NFAT signaling involving DYRK1A by

imaging the translocation of Flag-tagged NFATc1. Flag-NFATc1remained predominantly in the cytosol when expressed in 293Tcells (Mock, Fig. 5A). Upon the addition of the calcium ionophoreionomycin (IM), Flag-NFATc1 was translocated into the nucleus(Fig. 5A, IM) as a result of NFATc1 dephosphorylation by activatedcalcineurin. Overexpression of DYRK1A relocalized Flag-NFATc1into the cytoplasm, even in the presence of ionomycin owing to theopposing action of DYRK1A on NFATc1 phosphorylation, whichis consistent with its negative regulatory role in the calcineurin-NFAT pathway (Fig. 5A, DYRK1A+IM). Under these conditions,10 μM of CX-4945 effectively induced the nuclear translocation ofFlag-NFATc1, even in the presence of overexpressed DYRK1A(Fig. 5A, DYRK1A+IM+CX-4945).

The effect of CX-4945 on NFAT signaling was also examinedwith an alternative assay that measures NFATc1-mediatedtranscriptional activation, utilizing a firefly luciferase reporterdriven by the NFAT response element (NRE-Luc). Treatment

Fig. 2. ATP-competitive inhibition of DYRK1A byCX-4945. (A) Lineweaver–Burk plots for ATP in thepresence of CX-4945 at various concentrations.Data are mean±s.d. (B) Structural model of DYRK1Acomplexed with CX-4945, which is predicted to bindto the ATP-binding site of DYRK1A. (C) Thepredicted binding mode of CX-4945 to the ATP-binding pocket of DYRK1A. Amino acids in the ATP-binding pocket of DYRK1A are denoted by graysticks, and CX-4945 is indicated by yellow sticks.Hydrogen bonds are denoted as dotted black lines,and oxygen, nitrogen and chlorine atoms areindicated as red, blue and green sticks, respectively.

Fig. 3. CX-4945 potently inhibits DYRK1A-mediated Tau phosphorylation in mammalian cells. (A) Tau phosphorylation by DYRK1A in mammalian cellsis inhibited by CX-4945. 293T cells were transfected with plasmids expressing Tau and DYRK1A, and then treated with the indicated doses of CX-4945 for 6 h.Total cell extracts were prepared and subjected to western blotting with anti-Tau, anti-phosphorylated-Tau (at T212, p-Tau) and anti-DYRK1A antibodies. ThehnRNP A1 and GAPDH proteins were also monitored as controls. Western blotting was performed twice, and representative data are presented. (B) Thephosphorylated and total Tau proteins from western blotting were quantified, and the amount of protein relative to that in the DMSO-treated samples arepresented. The mean±s.d. was determined from two independent experiments. pTau/Tau, the ratio of phosphorylated Tau to total Tau. Vec, empty vector.

841

RESEARCH ARTICLE Disease Models & Mechanisms (2016) 9, 839-848 doi:10.1242/dmm.025668

Disea

seModels&Mechan

isms

Page 4: A chemical with proven clinical safety rescues Down ...A chemical with proven clinical safety rescues Down-syndrome-related phenotypes in through DYRK1A inhibition Hyeongki Kim1,2,*,

with IM and phorbol 12-myristate 13-acetate (PMA) induced adramatic increase in luciferase activity (∼20 fold) as a result ofthe nuclear translocation of NFATc1 and the subsequenttranscriptional activation (Fig. 5B). In this situation, theoverexpression of DYRK1A suppressed the stimulatory effect ofIM and PMA by 75%, resulting in a 4.5-fold increase inluciferase activity compared with that of the control (Fig. 5B).CX-4945 treatment reversed the effect of DYRK1Aoverexpression in a dose-dependent manner. The recovery effectwas initiated at low micromolar concentrations (1 and 3 μM),exponentially increased at 10 μM of CX-4945 and eventuallysurpassed the luciferase activity from cells that lacked exogenousDYRK1A expression (Fig. 5B). The overriding effect on NFATsignaling seen at 10 μM of CX-4945 might be explained by theinhibition of endogenous DYRK-family kinases in 293T cells.These results further confirm the inhibitory function of CX-4945on DYRK1A, particularly in DS- and AD-related signaling.

CX-4945 restores neurological and phenotypic defects in aDS- and AD-like Drosophila modelDrosophila melanogaster is a highly tractable genetic modelorganism that can be used to investigate the molecular mechanismsof human diseases. Basic biological and neurological propertiesare highly conserved between humans and Drosophila, and nearly75% of human disease-related genes, including DRYK1A, have afunctional homolog in the Drosophila genome. Based on theseadvantages, Drosophila has been frequently utilized as a powerfulin vivomodel for the pharmacological validation of therapeutic drugcandidates.

The Drosophila gene minibrain (mnb) is a well-studied orthologof human DYRK1A. Similar to DYRK1A in humans, mnb is highlyexpressed in neural tissues in Drosophila, and mutations in mnbcause phenotypic defects in neuroblast proliferation and braindevelopment. Similarly, the tissue-specific overexpression of mnbalso induces various phenotypic and neurological defects in

Fig. 4. CX-4945 inhibits DYRK1A-mediated APP and PS1phosphorylation in mammaliancells. 293T cells were transfectedwith the indicated plasmidsexpressing APP (A), PS1 (C) and/orDYRK1A. Transfected cells weretreated with the indicated doses ofCX-4945 for 6 h, and then total cellextracts were subjected to westernblot analysis. The phosphorylatedand total APP and PS1 proteins fromwestern blotting were quantified, andthe amounts of protein relative tothose of DMSO-treated samples areshown in B and D, respectively. Themean±s.d. values were determinedfrom two independent assays. p-APP,phosphorylated APP; p-PS1,phosphorylated PS1; Vec, emptyvector. / denotes the ratio of the twoindicated proteins.

Fig. 5. CX-4945 has a modulatory effect on DYRK1A-associated NFAT signaling. (A) The effect of CX-4945 on calcineurin-NFAT signaling wasanalyzed by visualizing the translocation of the NFATc1 protein. 293T cells were transfected with plasmids expressing Flag-NFATc1 or DYRK1A and then treatedwith CX-4945 (10 μM) or DMSO prior to stimulation with ionomycin (IM; 5 μM). (B) The effect of CX-4945 on NFATc1-mediated transcriptional activation. 293Tcells were transfected with NRE-Luc reporter plasmid with or without plasmid expressing DYRK1A, and cells were then treated with IM (5 μM) and phorbol12-myristate 13-acetate (PMA; 10 nM) along with the indicated doses of CX-4945. Firefly luciferase activities were measured using One-Glo reagents. Luciferaseactivity in the sample with reporter plasmid alone was set to 1, and the relative luciferase activities were calculated. Means±s.d. were determined from twoindependent experiments.

842

RESEARCH ARTICLE Disease Models & Mechanisms (2016) 9, 839-848 doi:10.1242/dmm.025668

Disea

seModels&Mechan

isms

Page 5: A chemical with proven clinical safety rescues Down ...A chemical with proven clinical safety rescues Down-syndrome-related phenotypes in through DYRK1A inhibition Hyeongki Kim1,2,*,

peripheral tissue, such as wings, as well as in the central nervoussystem (CNS) structure, recapitulating DS-like phenotypes in theDrosophila system (Degoutin et al., 2013).In order to validate the pharmacological use of CX-4945 in an

in vivo system, we established mnb-overexpressing flies andcharacterized the accompanying neurological and phenotypicdefects. The most prominent phenotypic defect was observed inthe morphology of the wing. Similar to in the report by Degoutinet al. (2013), wing-tissue-specific overexpression of mnb by the useof the MS1096-gal4 driver caused incomplete formation of lateralvein 5 (LV5) with a penetration of 90% (Fig. S3). Surprisingly,feeding newly hatched larvae with CX-4945 for 2 weeksconsiderably improved the wing defect. This phenotypic rescuewas seen when using each of the applied doses (1-25,000 nM) andwas even apparent with nanomolar concentrations of CX-4945 (10-100 nM), whereas much higher concentrations of harmine (10-25 μM) were required to achieve a similar effect (Fig. S3). Thisresult is consistent with our other observations that CX-4945 hasstronger inhibitory effects compared to harmine (Figs 1 and 3;Figs S1-S3). Moreover, these results suggest that 100 nM of CX-4945 is an optimal concentration for further examination in anmnb-overexpressing Drosophila system.As shown in Fig. 3A, Tau phosphorylation induced by DYRK1A

overexpression was significantly decreased by CX-4945 inmammalian cells. Similarly, to test the pharmacological effects of

CX-4945 on a Tau-mediated phenotype in the Drosophila model,mnb was overexpressed in eye tissue with or without the human Tauprotein, using the UAS-Gal4 system. The expression of human Tauin Drosophila retinal tissue (GMR>Tau) produced a small rougheye phenotype (GMR>Tau in Fig. 6A and B), as describedpreviously (Jackson et al., 2002). Coexpression of mnb enhancedTau toxicity and resulted in a more severe phenotype with smallereyes (GMR>mnb+Tau in Fig. 6A and B). Consistent with theinhibitory effect of CX-4945 on DYRK1A-mediated Tauphosphorylation in 293T cells (Fig. 3), eye defects induced bythe overexpression of Tau alone (GMR>Tau) or with mnb(GMR>mnb+Tau) were significantly mitigated by feeding newlyhatched larvae with 100 nM of CX-4945 [GMR>Tau (CX-4945)andGMR>mnb+Tau (CX-4945) in Fig. 6A and B]. Next, in order toanalyze the effect of CX-4945 on the neurological phenotype,primary neuron clusters and their primary axon bundles werelabeled with UAS-Synaptobrevin-GFP, driven by elav-Gal4. Pan-neuronal mnb overexpression (elav::synGFP>mnb) caused severeneurogenic defects in both the central and peripheral nervous systemduring embryogenesis [elav::synGFP>mnb (DMSO) in Fig. 6C].Feeding parent flies with 100 nM of CX-4945 restored theseneurogenic defects, particularly in the peripheral nervous system[elav::synGFP>mnb (CX-4945) in Fig. 6C]. It is noteworthy thatCX-4945 itself had little noticeable effect on control flies thatlacked mnb overexpression [elav::synGFP>control (CX-4945) in

Fig. 6. CX-4945 rescues neurological and phenotypic defects in anmnb-overexpressingDrosophilamodel. (A) The eyes of adult flies overexpressingmnband/or human Tau under the control of the eye-specificGMR-Gal4 driver, and of control flies bearing onlyGMR-Gal4. Overexpression ofmnb aggravated the eyeabnormality induced by Tau overexpression. This eye defect was effectively rescued by feeding with 100 nM of CX-4945. White dashed lines outline the eyecontour. (B) The retinal surface areas were measured on multiple samples (n>10) from each genotype, and average eye sizes were presented as normalizedpercentages of the DMSO-treatedGMR-Gal4 control. (C) The brain cortex, primary neuronal cell clusters and axon bundle, as visualized with GFP (green) usingUAS-Synaptobrevin-GFP driven by elav-Gal4 at the late embryonic stage 18. The pattern of neural connectivity and CNS structure were severely disorganized inmnb-overexpressing embryos (elav-synGFP>mnb, DMSO), the neurogenic defects of which were remarkably rescued by 100 nM of CX-4945. (D) The pan-neuronal overexpression ofmnb resulted in about 70% adult lethality. Feeding with 100 nM of CX-4945 dramatically reduced the lethality of the progeny by morethan 70% (n=100 for each group). The P-value is depicted with an asterisk (**P<0.01) (two-tailed Student’s t-test). Data are means±s.d. n.s., not significant.

843

RESEARCH ARTICLE Disease Models & Mechanisms (2016) 9, 839-848 doi:10.1242/dmm.025668

Disea

seModels&Mechan

isms

Page 6: A chemical with proven clinical safety rescues Down ...A chemical with proven clinical safety rescues Down-syndrome-related phenotypes in through DYRK1A inhibition Hyeongki Kim1,2,*,

Fig. 6C]. Moreover, the viability of adult flies with the neuronaloverexpression of mnb was examined. Pan-neuronal overexpressionofmnb caused high rates of mortality in adult flies (∼70%, Fig. 6D).Surprisingly, feeding parent flies with 100 nM of CX-4945dramatically reduced the mortality of the progeny by more than70%. Collectively, these results indicate that CX-4945 can functionas an inhibitor of DYRK1A/mnb in vivo, and can rescue theneurological and phenotypic defects of a DS- and AD-likeDrosophila model.

Oral administration of CX-4945 suppresses Tauhyperphosphorylation in the hippocampus of a DS-likemouseIn order to further validate the inhibition of DYRK1A by CX-4945in a higher-level organism, mice with three copies of the DYRK1Agene, which mimics the trisomy seen in DS, were used. This mousemodel has 50% higher expression of DYRK1A compared with anormal mouse, and increased Tau phosphorylation at T212 in thehippocampus has been well documented (Ryoo et al., 2007).Indeed, this phenomenon was also confirmed in our experiment. AllDYRK1A-overexpressing mice exhibited higher levels of Tauphosphorylation than those seen in normal mice (Fig. 7A and B).Surprisingly, acute oral administration (30 min) of CX-4945(75 mg/kg of body weight) remarkably decreased thephosphorylation of Tau (at T212), bringing it close to the levelsseen in normal mice (Fig. 7C and D). This strong inhibitory effect ofCX-4945 was still observed with samples harvested at 1 h after oraladministration (data not shown). Collectively, these results indicatethat CX-4945 efficiently penetrates the blood-brain barrier (BBB)and potently inhibits DYRK1A in the hippocampus of theDYRK1A transgenic mouse.

DISCUSSIONDYRK1A has been suggested recently to be a promising moleculartarget for disease-modifying treatments for DS, especially in regardsto the aspect of mental retardation. Even though a few compoundshave been discovered and developed as potent inhibitors ofDYRK1A, their therapeutic application is limited bypharmacological side effects. In this study, we identified a novelinhibitor of DYRK1A, CX-4945, which showed stronger inhibitoryactivity than harmine, INDY and proINDY, which are well-knownpotent inhibitors of DYRK1A. Moreover, CX-4945 significantlyrestored the neurological and phenotypic defects induced by theoverexpression of mnb, an ortholog of human DYRK1A, in aDrosophila model, validating the pharmacological inhibition ofDYRK1A by CX-4945 in vivo. Importantly, the DYRK1Ainhibitory activity of CX-4945 was also proven in a DS-likemouse model through the dramatic suppression of Tauhyperphosphorylation.

Our extensive analysis using various assays clearly demonstratesthat CX-4945 directly inhibits DYRKs with a high potency. First,CX-4945 inhibited all DYRKs tested in an in vitro kinase assay,showing the strongest inhibitory effect with DYRK1A andDYRK1B (IC50=∼6 nM for both, Fig. 1B). Using the same assay,the inhibitory activity of harmine (IC50=∼100 nM for DYRK1A),an alkaloid obtained from plants and a widely used inhibitor ofDYRKs, was found to be about 20-fold weaker than that of CX-4945 (IC50=6.8 nM for DYRK1A) (compare Fig. 1B and C).Moreover, in a previous report, INDY and proINDY, which arebenzothiazole derivatives identified as strong inhibitors ofDYRK1A, showed slightly weaker inhibitory activities thanharmine (Ogawa et al., 2010). Taken together, it appears that CX-4945 is more potent than harmine, INDY and proINDY. Second,

Fig. 7. Oral administration of CX-4945 suppresses Tau phosphorylation in DYRK1A-overexpressing mice. (A) Hippocampuses from normal andDYRK1A-overexpressing C57BL/6 mice (n=5 for each) were harvested, and the phosphorylation of Tau was analyzed by western blotting with anti-phosphorylated-Tau (at residue T212; p-Tau) and anti-Tau antibodies. α-Tubulin was also analyzed as a loading control. (B) The phosphorylated and total Tauproteins in panel A were quantified, and the mean amount of each protein was calculated. Relative ratios of phosphorylated Tau to total Tau (pTau/Tau) werepresented by setting the value from normal mice as 100%. (C) DYRK1A-overexpressing C57BL/6 mice (n=5 for each group) were administered orally with DMSOor 75 mg/kg of body weight of CX-4945 in PBS. Thirty minutes after oral administration, mice were killed, and the hippocampus was dissected from each mouse.Total cell lysates were prepared and subjected to western blotting with the indicated antibodies. α-Tubulin was also analyzed as a loading control. (D) Thephosphorylated and total Tau proteins in panel C were quantified, and the mean amount of each protein across the animals was calculated. Relative ratios ofphosphorylated Tau to total Tau were calculated by setting the value from DYRK1A mice that had been treated with DMSO as 100%. Data are means±s.d.(two-tailed Student’s t-test).

844

RESEARCH ARTICLE Disease Models & Mechanisms (2016) 9, 839-848 doi:10.1242/dmm.025668

Disea

seModels&Mechan

isms

Page 7: A chemical with proven clinical safety rescues Down ...A chemical with proven clinical safety rescues Down-syndrome-related phenotypes in through DYRK1A inhibition Hyeongki Kim1,2,*,

stronger inhibition of DYRK1A by CX-4945 was also confirmed inmammalian cells through the analysis of DYRK1A-mediated Tauphosphorylation. The estimated IC50 of CX-4945 (∼100-200 nM)on Tau phosphorylation that had been induced by DYRK1Aoverexpression was lower than that of harmine, INDY and proINDY(∼500, 2000 and 2000 nM, respectively; compare Fig. 3 and Fig.S1). Third, NFATc1-dependent transcriptional activation incalcineurin-NFAT signaling suppressed by the overexpression ofDYRK1A was dose-dependently derepressed by treatment withCX-4945, harmine, INDY and proINDY. However, CX-4945exhibited a stronger effect than harmine at the same concentration(Fig. S2). Fourth, a stronger effect of CX-4945 was observed inphenotype-rescue experiments using an mnb-overexpressingDrosophila model (Fig. S3). Notably, the oral administration ofCX-4945 dramatically suppressed the phosphorylation of Tau in thehippocampus of DS-like micewithin 30 min (Fig. 7), demonstratingits efficient penetrability through the BBB and DYRK1A-inhibitoryactivity in a higher-animal model. Collectively, these results firmlydemonstrate that CX-4945 is a potent inhibitor of DYRK1A in vitro,in mammalian cells and in living organisms.Intriguingly, the chemical structure of CX-4945 is quite similar

to that of harmine, except that CX-4945 has an additionalsubstructure appended to the central tricycle (Fig. S4). It isconceivable that this appendage might contribute considerably tothe enhanced inhibitory activity of CX-4945, considering that theinhibitory activity of CX-4945 is about threefold stronger thanthat of harmine in mammalian cells and 20-fold stronger in vitro(Figs 1 and 3). INDY and proINDY also have some degree ofsimilarity to the core structure of CX-4945 (Fig. S4). Consistentwith the similarity in chemical structures, the predicted structure ofthe CX-4945-DYRK1A complex is quite similar to co-crystalstructures of the harmine- and INDY-DYRK1A complexes.Determination of the co-crystal structure of CX-4945-DYRK1Ain a future study would improve our understanding of the molecularbasis of inhibition.Including harmine, INDY and proINDY, currently available

DYRK1A inhibitors can be classified into two categories: naturaland synthetic products. Harmine and epigallocatechin gallate (EGCG)are well-studied natural compounds (Smith et al., 2012). INDY,proINDY, roscovitine, purvalanol A, pyrazolidine-diones, amino-quinazolines, meridianins, pyridine and pyrazines, andchromenoidoles are all synthetic compounds with varying potenciestowards DYRK1A (Smith et al., 2012). Among these compounds,harmine, INDY and proINDY have drawn particular attention becauseof their high potency and selectivity. Harmine is currently consideredto be a potent inhibitor, but behavioral side effects related tomonoamine oxidase A (MOA) inhibition and hallucinogenicstimulation in animal models have limited its therapeutic application(Kim et al., 1997). According to a report byOgawa et al. (2010), INDYand its acetylated analogue proINDY are a little less potent but moreselective than harmine. Importantly, the in vivo use of proINDY hadbeen partly validated by showing the considerable rescue ofneurological and morphological defects induced by theoverexpression of Xenopus DYRK1A during development ofXenopus embryos. However, further extensive pharmacologicalvalidation in higher-level organisms is required. In addition, EGCG,a well-known phenolic compound from green tea, is of particularinterest given that beneficial effects on cognitive defects in aDSmousehas been observed (De la Torre et al., 2014). Even thoughEGCG is thesole compound that has been pharmacologically validated in an animalmodel, the nonselective nature and low potency of EGCG remains tobe tested further in humans in a clinical setting. Extensive evaluation of

leading or the most advanced candidate compounds suggests thatundesirable side effects are the limiting factors for their therapeuticapplication. In that sense, CX-4945, a newly identified inhibitor ofDYRK1A, would hold promise because its safety has been alreadyproven in phase I clinical trials for use as a cancer therapeutic. To ourknowledge, CX-4945 is the only drug candidate under clinicalinvestigation for which the DYRK1A-inhibitory effect has beenproven. Currently, phase 1b and phase 2 studies are underway incombination with gemcitabine and cisplatin for the frontline treatmentof individuals with bile duct cancers (cholangiocarcinoma), accordingto a release from Senhwa Biosciences (2014).

In addition to its significance in DS, the overexpression orhyperactivity of DYRK1A has been also reported in the brains ofindividuals diagnosed with any one of several neurodegenerativediseases, including AD, Parkinson’s, Huntington’s and Pick’s(Ferrer et al., 2005; Kang et al., 2005; Kim et al., 2006). Inparticular, the central role of DYRK1A in the pathogenesis of ADhas been well documented in a growing body of evidence. First,most DS individuals over 30 years old develop amyloid plaques andNFTs, which are the typical neuropathologies observed in the brainsof individuals with AD, leading to the progressiveneurodegeneration and high incidences of dementia at older ages(Dierssen and de Lagran, 2006; Wegiel et al., 2011). Second,DYRK1A directly phosphorylates Tau as well as APP and PS1, andaberrant and/or excessive phosphorylation of these targets isbelieved to be crucial for the formation of NFTs and amyloidplaques, respectively (Ryoo et al., 2008, 2007; Ryu et al., 2010).These observations provide the molecular link between DS and AD,and further suggest that DYRK1A is a promising therapeutic targetfor treating diseases such as AD and DS. In this regard, CX-4945,with a high potency and proven safety in a clinical setting, will be auseful tool in DYRK1A-related research and can serve as a drugcandidate for the treatment of DS and AD.

In addition to neurodegenerative diseases, DYRK1A has alsobeen reported to be associated with cancers and metabolic diseases.DYRK1A is involved in the progression of cancer by modulatingseveral pathways related to proliferation and migration (Ionescuet al., 2012). The involvement of DYRK1A in metabolic events hasbeen demonstrated particularly by our study with mnb, an orthologof human DYRK1A, in the Drosophila model. Overexpressionof mnb in peptidergic neurons results in increased food intake(Hong et al., 2012). Notably, a recent study has highlighted thesignificance and versatility of DYRK1A by showing that inhibitionof DYRK1A by harmine increases human pancreatic β-cellproliferation through the modulation of NFAT signaling (Wanget al., 2015), indicating a new therapeutic approach for diabetes.Therefore, targeting DYRK1A would be broadly applicable as atherapeutic approach towards at least some types of cancers andmetabolic diseases.

Our research suggests that CX-4945, a drug candidate for cancertreatment, can be repurposed for disease-modifying treatment of DSand AD. Further pharmacological validation in higher-animalmodels for DS and AD is required in future studies.

MATERIALS AND METHODSCell culture, drug treatment and transfection293T cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM)containing 10% fetal bovine serum (FBS; Hyclone) supplemented with 1%penicillin and streptomycin. CX-4945 (Selleckchem), harmine (Sigma),INDY and proINDY (Tocris) were dissolved in dimethyl sulfoxide (DMSO)prior to treatment. Cells were seeded at approximately 50% confluence into6-well cell culture plates, maintained overnight and then transfected with the

845

RESEARCH ARTICLE Disease Models & Mechanisms (2016) 9, 839-848 doi:10.1242/dmm.025668

Disea

seModels&Mechan

isms

Page 8: A chemical with proven clinical safety rescues Down ...A chemical with proven clinical safety rescues Down-syndrome-related phenotypes in through DYRK1A inhibition Hyeongki Kim1,2,*,

appropriate plasmids using the XtremeGene Transfection Reagent (Roche)according to the manufacturer’s instructions.

Plasmid constructionPcDNA3.1-DYRK1A, -Tau, -APP and -C-PS1 were constructed aspreviously described (Ryoo et al., 2008, 2007; Ryu et al., 2010). Togenerate a plasmid expressing the Flag-NFATc1 protein, NFATC1 cDNAwas amplified by using PCR with appropriate primers, and the DNAfragment was inserted into the EcoRI site of the pcDNA3.1-Flag plasmid.

Quantitative western blot analysisQuantitative western blot analysis was performed as described previously(Choi et al., 2014). The following antibodies were used: anti-Tau antibody(HT7, catalog # MN1000, ThermoFisher Scientific, 1:1000 dilution), anti-phosphorylated-Tau (at residue T212) antibody (catalog # 44740G,Invitrogen, 1:1000 dilution), anti-glyceraldehyde-phosphate-dehydrogenase(GAPDH) antibody (G-9, catalog # sc-365062, Santa Cruz Biotechnology,1:2000 dilution), anti-PS1 antibody, (catalog # 3622, Cell SignalingTechnology) as well as anti-APP, anti-phospho-APP (at residue T668) andanti-Flag antibodies (catalog # 2452, catalog # 2451, catalog # 9146, all CellSignaling Technology, 1:1000, 1:1000 and 1:1000 dilution, respectively).Anti-DYRK1A antibody (1:500 dilution) was generated as describedpreviously (Ryoo et al., 2007). Anti-hnRNP-A1 antibody (1:1000 dilution)was kindly provided by Gideon Dreyfuss (University of Pennsylvania, PA).

In vitro kinase assaysKinase assays were conducted using the Kinase Profiler services offered byLife Technologies, which utilizes a fluorescence-based immunoassay.Detailed protocols can be found at http://www.lifetechnologies.com/kinaseprofiling. The inhibitory activity of each kinase (DYRK1A,DYRK1B, DYRK3 and DYRK4) was measured with five concentrationsof CX-4945 over a range of 0.001 to 10 μM, and the IC50 was determinedusing the GraphPad Prism 5 software. To determine whether CX-4945 actsby competing with ATP for the inhibition of DYRK1A, kinase activity wasmeasured in the presence of various concentrations of ATP (50, 75, 100, 150and 200 μM), and IC50 values were determined using the GraphPad Prism 5software. All experiments were performed twice.

Computer-aided molecular dockingTo build a structural model of DYRK1A in complex with CX-4945, weperformedmolecular docking studies by using the CDOCKERprogram (Wuet al., 2003) in Discovery Studio 3.1 software (Accelrys). The CX-4945ligand was generated by using ChemBioDraw, and the energy-minimizedstructure was transferred to Discovery Studio 3.1. The crystal structure ofDYRK1A obtained from the RCSB Protein Data Bank (PDB code: 3ANQ)was used for docking studies. Ten initial random conformations were taken,and ten initial poses were chosen for final refinement. CHARMm forcefieldwas employed for molecular dynamics and dynamics steps, and targettemperatureswere set to be 1000 and 1000 K, respectively. The structurewiththe lowest CDOCKER interaction energy was selected to be a final structure.Figures of the complex were drawn by using the PyMOL software package.

NFATc1 localization by immunofluorescence293T cells were seeded into 8-well chambered coverslips (Ibidi) andtransfected with a plasmid expressing Flag-NFATc1. On the next day, cellswere pre-treated with CX-4945 (10 μM) for 3 h and then stimulated with IM(5 μM) for 1 h. Cells were fixed with 4% paraformaldehyde in PBS for10 min at room temperature, followed by permeabilization with 0.1% TritonX-100, and then subjected to immunofluorescence staining with anti-Flag(1:500) antibody for 2 h. Cells were then washed with cold PBS three timesfor 3 min each, incubated with Alexa-Fluor-488-conjugated anti-rabbit IgG(1:800) (Invitrogen) at room temperature for 1 h and then examined under afluorescence microscope.

NFAT-RE promoter assayThe firefly luciferase reporter assay was performed using a One-Glo AssaySystem (Promega) according to the manufacturer’s instructions. Thetranscriptional activity of NFATc1 in 293T cells was measured with a

reporter vector (pGL4.30 luc2P/NFAT-RE/Hygro) (Promega) thatexpresses the firefly luciferase gene under the control of an NFAT-REpromoter. 293T cells were transfected with the DYRK1A-expressingplasmid and reporter plasmid. On the next day, cells were treated with IM(2.5 μM) and phorbol-12-myristate-13-acetate (10 nM) along with theindicated amounts of inhibitors. Cells were further incubated for 8 h andthen cell lysates were harvested to measure luciferase activity.

Drosophila culture and drug administrationDrosophila melanogaster were cultured at 25°C on standard cornmealmedium. GMR-gal4 (#9146), MS1096-gal4 (#8860), elav[C155]-gal4::UAS-synGFP (#6923), UAS-Tau (human wild-type Tau, #51362) and allother stocks and balancers were obtained from the Bloomington StockCenter (Bloomington, IN).UAS-mnb transgenic flies have been described inour previous reports (Hong et al., 2012); the full-length coding sequence ofDrosophila mnb (CG 42273) was subcloned into the pUAS vector, andUAS-mnb transgenic flies were obtained by using the P-element-mediatedgermline transformation. For defining optimal concentration of chemicalinhibitors, 50 embryos of each genotype were raised on drug-containingfood (0, 1 nM, 10 nM, 100 nM, 1 µM, 10 µM and 25 µM in 0.05% DMSO)and allowed to develop to adulthood (viability assay and wing veinquantification assay). Experiments were repeated at least three times. Foranalyzing the eye phenotype, newly eclosed flies were collected and allowedto mate for 2-3 days and then transferred to medium containing 100 nMCX-4945 or DMSO as a control. Flies were exposed to the drug until adult stage,and the eye phenotypes of newly eclosed male were photographed using theOlympus SZ60 binocular microscope equipped with an eXcope K5CCD system. Fly eye sizes were measured on multiple samples (n>10)from each genotype using the National Institutes of Health ImageJ software.Average eye size was presented as a normalized percentage of control eyesize.

For analyzing the embryonic neurological phenotypes and lethality,newly eclosed virgin females or male flies were raised on standard mediumcontaining 0.5% DMSO or 100 nM CX-4945 for 7 days before mating.CX4945-exposed virgins and males were allowed to mate in drug-containing medium. Embryos from CX-4945-fed parent flies werecollected and staged at 25°C. They were dechorionated and fixed, andimmunostaining was performed as described previously (Lee et al., 2008).The samples were then rinsed and washed three times for 15 min each timein PBT (PBS, 0.1% Triton X-100) and blocked for 1 h in PBT plus 10%normal goat serum. Primary antibodies were incubated overnight at 4°C.Then, samples were washed and incubated in secondary antibodies for 2 h atroom temperature, washed again and mounted in Vectashield (VectorLaboratories, Burlingame, CA). The primary antibody used was chickenanti-GFP (Abcam, catalog #ab13970, 1:10,000), and Alexa-Fluor-488-coupled secondary antibodies were from Molecular Probes. Embryonicneurological phenotypes were imaged on an Olympus FV1000 confocalmicroscope (Olympus, Center Valley, PA) and processed using ImageJ. Foradult lethality recordings, at least 100 flies were used for each individualexperiment. For paired samples, two-tailed Student’s t-test was used toanalyze the effects of CX-4945 on eye phenotype and adult lethality.

Tau phosphorylation in the hippocampus of DYRK1A TG mouseDYRK1A transgenic mice with three copies of the human DYRK1A genewere maintained as described previously (Ahn et al., 2006). Experimentswere performed in accordance with the guidelines under the approval of theInstitutional Review Committee for Animal Care and Use, KRIBB,Daejeon, Korea. The 8- to 10-week-old C57BL/6 mice were administeredwith DMSO or CX-4945 (75 mg/kg of body weight) orally in PBSsolution, and the hippocampus was dissected after 30 min. Thehippocampal lysates were prepared by using a Digital Sonifier CellDisruptor instrument (BRANSON) in CelLytic MT buffer (catalog #C3228, Sigma-Aldrich) containing protease inhibitor cocktail set III(catalog # 535140-1ML, Calbiochem). The protein concentration wasdetermined using the Bradford protein assay (Bio-Rad). Proteins wereseparated by SDS-PAGE and analyzed by immunoblotting with antibodiesagainst phosphorylated Tau and Tau using an LAS4000 image analyzer(Fujifilm, Tokyo, Japan).

846

RESEARCH ARTICLE Disease Models & Mechanisms (2016) 9, 839-848 doi:10.1242/dmm.025668

Disea

seModels&Mechan

isms

Page 9: A chemical with proven clinical safety rescues Down ...A chemical with proven clinical safety rescues Down-syndrome-related phenotypes in through DYRK1A inhibition Hyeongki Kim1,2,*,

Statistical analysisData were analyzed by two-tailed paired or unpaired Student’s t-tests. *, **and *** represent P<0.05, P<0.01 and P<0.001, respectively, which areconsidered statistically significant.

AcknowledgementsWe thank Dr. Gideon Dreyfuss at University of Pennsylvania for kindly providing ananti-hnRNP-A1 antibody.

Competing interestsThe authors declare no competing or financial interests.

Author contributionsS.C., K.-S.L. and H.K. conceived the study. S.C., K.-S.L., S.-W.C., M.C. and H.K.designed the experiments. H.K., K.-S.L., A.-K.K., M.C., K.C., M.K., J.-S.L. andM.-S.L. performed the experiments. S.C., K.-S.L., S.-W.C., H.K., A.-K.K., M.C., K.C.and M.-S.L. analyzed the data. W.-J.S. and K.Y. provided valuable materials(DYRK1A transgenic mouse and mnb flies, respectively). S.C., K.-S.L., S.-W.L., H.K., J.-S.L. and S.-Y.L. wrote the manuscript. All authors read and approved the finalmanuscript.

FundingThis research was supported by a grant of the Korea Health Technology R&DProject through the Korea Health Industry Development Institute (KHIDI), funded bythe Ministry of Health and Welfare [grant numbers HI14C2124 and A092004]; theNational Research Foundation of Korea [grant numbers NRF-2014M3A9D8034462and NRF-2015M3A9C7030128]; and the Korea Research Institute of Bioscienceand Biotechnology Research Initiative Programs.

Supplementary informationSupplementary information available online athttp://dmm.biologists.org/lookup/doi/10.1242/dmm.025668.supplemental

ReferencesAdayev, T., Wegiel, J. and Hwang, Y.-W. (2011). Harmine is an ATP-competitiveinhibitor for dual-specificity tyrosine phosphorylation-regulated kinase 1A(Dyrk1A). Arch. Biochem. Biophys. 507, 212-218.

Ahn, K.-J., Jeong, H. K., Choi, H.-S., Ryoo, S.-R., Kim, Y. J., Goo, J.-S., Choi,S.-Y., Han, J.-S., Ha, I. and Song, W.-J. (2006). DYRK1A BAC transgenic miceshow altered synaptic plasticity with learning and memory defects.Neurobiol. Dis.22, 463-472.

Altafaj, X., Dierssen, M., Baamonde, C., Marti, E., Visa, J., Guimera, J., Oset, M.,Gonzalez, J. R., Florez, J., Fillat, C. et al. (2001). Neurodevelopmental delay,motor abnormalities and cognitive deficits in transgenic mice overexpressingDyrk1A (minibrain), a murine model of Down’s syndrome. Hum. Mol. Genet. 10,1915-1923.

Aranda, S., Laguna, A. and de la Luna, S. (2011). DYRK family of protein kinases:evolutionary relationships, biochemical properties, and functional roles. FASEB J.25, 449-462.

Arque, G., Fotaki, V., Fernandez, D., Martinez de Lagran, M., Arbones, M. L. andDierssen, M. (2008). Impaired spatial learning strategies and novel objectrecognition in mice haploinsufficient for the dual specificity tyrosine-regulatedkinase-1A (Dyrk1A). PLoS ONE 3, e2575.

Arron, J. R., Winslow, M. M., Polleri, A., Chang, C.-P., Wu, H., Gao, X., Neilson,J. R., Chen, L., Heit, J. J., Kim, S. K. et al. (2006). NFAT dysregulation byincreased dosage of DSCR1 and DYRK1A on chromosome 21. Nature 441,595-600.

Becker, W. and Joost, H.-G. (1999). Structural and functional characteristics ofDyrk, a novel subfamily of protein kinases with dual specificity. Prog. Nucleic AcidRes. Mol. Biol. 62, 1-17.

Benavides-Piccione, R., Dierssen, M., Ballesteros-Yanez, I., Martinez deLagran, M., Arbones, M. L., Fotaki, V., DeFelipe, J. and Elston, G. N. (2005).Alterations in the phenotype of neocortical pyramidal cells in the Dyrk1A+/-mouse. Neurobiol. Dis. 20, 115-122.

Choi, K., Kim, H., Kang, H., Lee, S.-Y., Lee, S. J., Back, S. H., Lee, S. H., Kim,M. S., Lee, J. E., Park, J. Y. et al. (2014). Regulation of diacylglycerolacyltransferase 2 protein stability by gp78-associated endoplasmic-reticulum-associated degradation. FEBS J. 281, 3048-3060.

De la Torre, R., De Sola, S., Pons, M., Duchon, A., de Lagran, M. M., Farre, M.,Fito, M., Benejam, B., Langohr, K., Rodriguez, J. et al. (2014).Epigallocatechin-3-gallate, a DYRK1A inhibitor, rescues cognitive deficits inDown syndromemousemodels and in humans.Mol. Nutr. FoodRes. 58, 278-288.

Debdab, M., Carreaux, F., Renault, S., Soundararajan, M., Fedorov, O.,Filippakopoulos, P., Lozach, O., Babault, L., Tahtouh, T., Baratte, B. et al.(2011). Leucettines, a class of potent inhibitors of cdc2-like kinases and dualspecificity, tyrosine phosphorylation regulated kinases derived from the marine

sponge leucettamine B: modulation of alternative pre-RNA splicing. J. Med.Chem. 54, 4172-4186.

Degoutin, J. L., Milton, C. C., Yu, E., Tipping, M., Bosveld, F., Yang, L.,Bellaiche, Y., Veraksa, A. and Harvey, K. F. (2013). Riquiqui and minibrain areregulators of the hippo pathway downstream of Dachsous. Nat. Cell Biol. 15,1176-1185.

Dierssen, M. and de Lagran, M. M. (2006). DYRK1A (dual-specificity tyrosine-phosphorylated and -regulated kinase 1A): a gene with dosage effect duringdevelopment and neurogenesis. ScientificWorldJournal 6, 1911-1922.

Fedorov, O., Huber, K., Eisenreich, A., Filippakopoulos, P., King, O., Bullock,A. N., Szklarczyk, D., Jensen, L. J., Fabbro, D., Trappe, J. et al. (2011). SpecificCLK inhibitors from a novel chemotype for regulation of alternative splicing.Chem.Biol. 18, 67-76.

Ferguson, A. D., Sheth, P. R., Basso, A. D., Paliwal, S., Gray, K., Fischmann,T. O. and Le, H. V. (2011). Structural basis of CX-4945 binding to human proteinkinase CK2. FEBS Lett. 585, 104-110.

Ferrer, I., Barrachina, M., Puig, B., Martinez de Lagran, M., Marti, E., Avila, J.and Dierssen, M. (2005). Constitutive Dyrk1A is abnormally expressed inAlzheimer disease, Down syndrome, Pick disease, and related transgenicmodels. Neurobiol. Dis. 20, 392-400.

Fotaki, V., Dierssen, M., Alcantara, S., Martinez, S., Marti, E., Casas, C., Visa, J.,Soriano, E., Estivill, X. and Arbones, M. L. (2002). Dyrk1A haploinsufficiencyaffects viability and causes developmental delay and abnormal brain morphologyin mice. Mol. Cell. Biol. 22, 6636-6647.

Fotaki, V., Martinez De Lagran, M., Estivill, X., Arbones, M. and Dierssen, M.(2004). Haploinsufficiency of Dyrk1A in mice leads to specific alterations in thedevelopment and regulation of motor activity. Behav. Neurosci. 118, 815-821.

Guimera, J., Casas, C., Estivill, X. and Pritchard, M. (1999). Human minibrainhomologue (MNBH/DYRK1): characterization, alternative splicing, differentialtissue expression, and overexpression in Down syndrome. Genomics 57,407-418.

Hong, S.-H., Lee, K.-S., Kwak, S.-J., Kim, A.-K., Bai, H., Jung, M.-S., Kwon, O.-Y.,Song, W.-J., Tatar, M. and Yu, K. (2012). Minibrain/Dyrk1a regulates food intakethrough the Sir2-FOXO-sNPF/NPY pathway in Drosophila and mammals. PLoSGenet. 8, e1002857.

Ionescu, A., Dufrasne, F., Gelbcke, M., Jabin, I., Kiss, R. and Lamoral-Theys, D.(2012). DYRK1A kinase inhibitors with emphasis on cancer. Mini Rev. Med.Chem. 12, 1315-1329.

Jackson, G. R., Wiedau-Pazos, M., Sang, T.-K., Wagel, N., Brown, C. A.,Massachi, S. and Geschwind, D. H. (2002). Human wild-type tau interacts withwingless pathway components and produces neurofibrillary pathology indrosophila. Neuron 34, 509-519.

Johnson, G. V. and Hartigan, J. A. (1999). Tau protein in normal and Alzheimer’sdisease brain: an update. J. Alzheimers Dis. 1, 329-351.

Kang, J. E., Choi, S. A., Park, J. B. and Chung, K. C. (2005). Regulation of theproapoptotic activity of huntingtin interacting protein 1 by Dyrk1 and caspase-3 inhippocampal neuroprogenitor cells. J. Neurosci. Res. 81, 62-72.

Kannan, N. and Neuwald, A. F. (2004). Evolutionary constraints associated withfunctional specificity of the CMGC protein kinases MAPK, CDK, GSK, SRPK,DYRK, and CK2alpha. Protein Sci. 13, 2059-2077.

Kim, H., Sablin, S. O. and Ramsay, R. R. (1997). Inhibition of monoamine oxidaseA by beta-carboline derivatives. Arch. Biochem. Biophys. 337, 137-142.

Kim, E. J., Sung, J. Y., Lee, H. J., Rhim, H., Hasegawa, M., Iwatsubo, T., Min,D. S., Kim, J., Paik, S. R. and Chung, K. C. (2006). Dyrk1A phosphorylatesalpha-synuclein and enhances intracellular inclusion formation. J. Biol. Chem.281, 33250-33257.

Kim, H., Choi, K., Kang, H., Lee, S.-Y., Chi, S.-W., Lee, M.-S., Song, J., Im, D.,Choi, Y. and Cho, S. (2014). Identification of a novel function of CX-4945 as asplicing regulator. PLoS ONE 9, e94978.

Lee, K.-S., Kwon, O.-Y., Lee, J. H., Kwon, K., Min, K.-J., Jung, S.-A., Kim, A.-K.,You, K.-H., Tatar, M. and Yu, K. (2008). Drosophila short neuropeptide Fsignalling regulates growth by ERK-mediated insulin signalling. Nat. Cell Biol. 10,468-475.

Mott, B. T., Tanega, C., Shen, M., Maloney, D. J., Shinn, P., Leister, W., Marugan,J. J., Inglese, J., Austin, C. P., Misteli, T. et al. (2009). Evaluation of substituted6-arylquinazolin-4-amines as potent and selective inhibitors of cdc2-like kinases(Clk). Bioorg. Med. Chem. Lett. 19, 6700-6705.

Ogawa, Y., Nonaka, Y., Goto, T., Ohnishi, E., Hiramatsu, T., Kii, I., Yoshida, M.,Ikura, T., Onogi, H., Shibuya, H. et al. (2010). Development of a novel selectiveinhibitor of the Down syndrome-related kinase Dyrk1A. Nat. Commun. 1, 86.

Ryoo, S.-R., Jeong, H. K., Radnaabazar, C., Yoo, J.-J., Cho, H.-J., Lee, H.-W.,Kim, I.-S., Cheon, Y.-H., Ahn, Y. S., Chung, S.-H. et al. (2007). DYRK1A-mediated hyperphosphorylation of Tau. A functional link betweenDown syndromeand Alzheimer disease. J. Biol. Chem. 282, 34850-34857.

Ryoo, S.-R., Cho, H.-J., Lee, H.-W., Jeong, H. K., Radnaabazar, C., Kim, Y.-S.,Kim, M.-J., Son, M.-Y., Seo, H., Chung, S.-H. et al. (2008). Dual-specificitytyrosine(Y)-phosphorylation regulated kinase 1A-mediated phosphorylation ofamyloid precursor protein: evidence for a functional link between Down syndromeand Alzheimer’s disease. J. Neurochem. 104, 1333-1344.

847

RESEARCH ARTICLE Disease Models & Mechanisms (2016) 9, 839-848 doi:10.1242/dmm.025668

Disea

seModels&Mechan

isms

Page 10: A chemical with proven clinical safety rescues Down ...A chemical with proven clinical safety rescues Down-syndrome-related phenotypes in through DYRK1A inhibition Hyeongki Kim1,2,*,

Ryu, Y. S., Park, S. Y., Jung, M.-S., Yoon, S.-H., Kwen, M.-Y., Lee, S.-Y., Choi,S.-H., Radnaabazar, C., Kim, M.-K., Kim, H. et al. (2010). Dyrk1A-mediatedphosphorylation of Presenilin 1: a functional link between Down syndrome andAlzheimer’s disease. J. Neurochem. 115, 574-584.

Siddiqui-Jain, A., Drygin, D., Streiner, N., Chua, P., Pierre, F., O’Brien, S. E.,Bliesath, J., Omori, M., Huser, N., Ho, C. et al. (2010). CX-4945, an orallybioavailable selective inhibitor of protein kinase CK2, inhibits prosurvival andangiogenic signaling and exhibits antitumor efficacy. Cancer Res. 70,10288-10298.

Smith, D. J. andRubin, E. M. (1997). Functional screening and complex traits: human21q22.2 sequences affecting learning in mice. Hum. Mol. Genet. 6, 1729-1733.

Smith, B., Medda, F., Gokhale, V., Dunckley, T. and Hulme, C. (2012). Recentadvances in the design, synthesis, and biological evaluation of selective DYRK1Ainhibitors: a new avenue for a disease modifying treatment of Alzheimer’s? ACSChem. Neurosci. 3, 857-872.

Tejedor, F. J. and Hammerle, B. (2011). MNB/DYRK1A as a multiple regulator ofneuronal development. FEBS J. 278, 223-235.

Tiraboschi, P., Hansen, L. A., Thal, L. J. and Corey-Bloom, J. (2004). Theimportance of neuritic plaques and tangles to the development and evolution ofAD. Neurology 62, 1984-1989.

Wang, P., Alvarez-Perez, J.-C., Felsenfeld, D. P., Liu, H., Sivendran, S., Bender,A., Kumar, A., Sanchez, R., Scott, D. K., Garcia-Ocana, A. et al. (2015). Ahigh-throughput chemical screen reveals that harmine-mediated inhibitionof DYRK1A increases human pancreatic beta cell replication. Nat. Med. 21,383-388.

Wegiel, J., Gong, C.-X. and Hwang, Y.-W. (2011). The role of DYRK1A inneurodegenerative diseases. FEBS J. 278, 236-245.

Wiseman, F. K., Alford, K. A., Tybulewicz, V. L. J. and Fisher, E. M. C. (2009).Down syndrome–recent progress and future prospects. Hum. Mol. Genet. 18,R75-R83.

Wu, G., Robertson, D. H., Brooks, C. L., III and Vieth, M. (2003). Detailed analysisof grid-based molecular docking: a case study of CDOCKER-A CHARMm-basedMD docking algorithm. J. Comput. Chem. 24, 1549-1562.

848

RESEARCH ARTICLE Disease Models & Mechanisms (2016) 9, 839-848 doi:10.1242/dmm.025668

Disea

seModels&Mechan

isms