Selectivity Profiling and Biological Activity of Novel β-Carbolines as Potent and Selective DYRK1 Kinase Inhibitors

Katharina Rüben, Anne Wurzlbauer, Agnes Walte, Wolfgang Sippl, Franz Bracher, Walter Becker, Katharina Rüben, Anne Wurzlbauer, Agnes Walte, Wolfgang Sippl, Franz Bracher, Walter Becker

Abstract

DYRK1A is a pleiotropic protein kinase with diverse functions in cellular regulation, including cell cycle control, neuronal differentiation, and synaptic transmission. Enhanced activity and overexpression of DYRK1A have been linked to altered brain development and function in Down syndrome and neurodegenerative diseases such as Alzheimer's disease. The β-carboline alkaloid harmine is a high affinity inhibitor of DYRK1A but suffers from the drawback of inhibiting monoamine oxidase A (MAO-A) with even higher potency. Here we characterized a series of novel harmine analogs with minimal or absent MAO-A inhibitory activity. We identified several inhibitors with submicromolar potencies for DYRK1A and selectivity for DYRK1A and DYRK1B over the related kinases DYRK2 and HIPK2. An optimized inhibitor, AnnH75, inhibited CLK1, CLK4, and haspin/GSG2 as the only off-targets in a panel of 300 protein kinases. In cellular assays, AnnH75 dose-dependently reduced the phosphorylation of three known DYRK1A substrates (SF3B1, SEPT4, and tau) without negative effects on cell viability. AnnH75 inhibited the cotranslational tyrosine autophosphorylation of DYRK1A and threonine phosphorylation of an exogenous substrate protein with similar potency. In conclusion, we have characterized an optimized β-carboline inhibitor as a highly selective chemical probe that complies with desirable properties of drug-like molecules and is suitable to interrogate the function of DYRK1A in biological studies.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. Inhibition of DYRK1A and related…
Fig 1. Inhibition of DYRK1A and related kinases by selected β-carbolines.
Kinase activities are given as the means of at least 3 measurements in the presence of 1 μM of the compounds (10 μM in HIPK2 assays) and are expressed as the percentage of the uninhibited control (Kinase-GLO assay). 5-iodotubercidin (IoT) served as a structurally unrelated control compound that inhibits all tested kinases.
Fig 2. Analysis of kinase inhibitor interactions.
Fig 2. Analysis of kinase inhibitor interactions.
A-D, Predicted binding modes of AnnH75 at DYRK1A and related kinases. The inhibitor is colored cyan and kinases are depicted as ribbon structures. Only relevant amino acid residues in the ATP binding pocket are shown for clarity. In B-D, residues different from DYRK1A are colored green and labeled. The distances of the two hydrogen bonds (red line) and the distance between the cyano group and the glycine of the P-loop (G166 in DYRK1A) are given in Angstrom. (DYRK1A, PDB ID 3ANR; DYRK2 PDB ID 4AZF; HIPK2, homology model, CLK1, PDB ID 2VAG). E, Comparison of amino acid residues relevant for AnnH75 binding. In the left, the relationship of the kinases is illustrated by the sequence identity of their catalytic domains. Residues generally conserved in protein kinases (G166, K188, D307) and the gatekeeper residue (F238) are highlighted by bold print. Residues different from DYRK1A are shown with their position in the sequence. Differences correlating with kinase resistance to AnnH75 are highlighted in red.
Fig 3. Selectivity profile of AnnH75.
Fig 3. Selectivity profile of AnnH75.
AnnH75 was profiled at a concentration of 1 μM against a panel of 300 protein kinases (see Table A in S1 File for the complete results). A, Target kinases inhibited by more than 50% are indicated. The kinome dendrogram was adapted and is reproduced courtesy of Cell Signaling Technology. B, Calculation of the Gini coefficient as a measure of kinase selectivity. The Lorenz curve illustrates the degree to which the total inhibitory activity of a compound (i.e. the sum of inhibition of all tested kinases) is equally distributed among all tested kinases (bisector line, Gini coefficient of 0) or directed towards a single kinase (maximal selectivity, a Gini coefficient of 1).
Fig 4. Inhibition of SF3B1 phosphorylation by…
Fig 4. Inhibition of SF3B1 phosphorylation by DYRK1A in HeLa cells.
HeLa cells expressing GFP-SF3B1-NT were treated with the indicated compounds for 18 h. The phosphorylation state of SF3B1 was determined by immunoblotting with pT434 antibody, and the results were normalized to the total amount of SF3B1 immunoreactivity. A, Representative western blots. AnnH79 is a harmine analogue that does not inhibit DYRK1A and was used as negative control. The vertical line indicates where irrelevant lanes were deleted from the final image. B, The column diagram summarizes the quantitative evaluation of 3–6 experiments for each compound (means + SD).
Fig 5. Inhibition of SEPT4 and tau…
Fig 5. Inhibition of SEPT4 and tau phosphorylation by DYRK1A.
A, HeLa cells transiently expressing FLAG-SEPT4 were treated with AnnH31 or AnnH75 for 5 h before cells were lysed and analysed by immunoblotting with a FLAG-tag antibody. 5-iodotubercidin (IoT) served as positive control. Relative SEPT4 phosphorylation was calculated as the ratio of the intensities of the phosphorylated upper band and the lower band. B, HEK293 cells with constitutive expression of GFP-tau and regulatable expression of GFP-DYRK1A were treated with doxycyclin and the indicated inhibitors for 18 h. Phosphorylation of tau on Thr212 was detected with a phosphospecific antibody. Expression levels of GFP-tau and GFP-DYRK1A were assessed with a GFP antibody. For quantitative evaluation of DYRK1A inhibition, the basal pT212 signal in control cells not treated with doxycyclin (Ctrl) was subtracted from all values. C, Quantitative evaluation of three experiments each for SEPT4 and tau. All data were standardized to the level of phosphorylation in cells untreated with inhibitors. Error bars indicate SEM.
Fig 6. AnnH75 inhibits both threonine and…
Fig 6. AnnH75 inhibits both threonine and tyrosine kinase activity of DYRK1A.
A DYRK1A construct with an N-terminal StrepTag 2 (ST2-DYRK1Acat) was expressed in a cell-free E. coli-derived expression system. Coupled in vitro transcription and translation reactions were incubated for 1 h in the presence of recombinant SF3B1-NT-His6 and AnnH75. Phosphorylation of tyrosines in DYRK1A and of Thr434 in SF3B1 was determined by immunoblotting with a phosphotyrosine-specific antibody (pTyr) and a pThr434-specific antibody. A, Representative western blots. The asterisks mark unidentified bands. B, Quantitative evaluation. Results were normalized to the total amount of DYRK1A or SF3B1, respectively, and are plotted relative to the phosphorylation in the untreated control samples (means +/- SEM, n = 3).

References

    1. Cohen P, Alessi DR. Kinase Drug Discovery—What's Next in the Field? ACS Chem Biol. 2013;8: 96–104. 10.1021/cb300610s
    1. Knapp S, Arruda P, Blagg J, Burley S, Drewry DH, Edwards A, et al. A public-private partnership to unlock the untargeted kinome. Nat Chem Biol. 2013;9: 3–6. 10.1038/nchembio.1113
    1. Knight ZA, Shokat KM. Features of selective kinase inhibitors. Chem Biol. 2005;12: 621–637.
    1. Cohen P. Guidelines for the effective use of chemical inhibitors of protein function to understand their roles in cell regulation. Biochem J. 2010;425: 53–54.
    1. Frye SV. The art of the chemical probe. Nat Chem Biol. 2010;6: 159–161.
    1. Workman P, Collins I. Probing the probes: fitness factors for small molecule tools. Chem Biol. 2010;17: 561–577. 10.1016/j.chembiol.2010.05.013
    1. Lochhead PA, Sibbet G, Morrice N, Cleghon V. Activation-loop autophosphorylation is mediated by a novel transitional intermediate form of DYRKs. Cell. 2005;121: 925–36.
    1. Becker W, Sippl W. Activation, regulation, and inhibition of DYRK1A. FEBS J. 2011;278: 246–56. 10.1111/j.1742-4658.2010.07956.x
    1. Walte A, Rüben K, Birner-Gruenberger R, Preisinger C, Bamberg-Lemper S, Hilz N, et al. Mechanism of dual specificity kinase activity of DYRK1A. FEBS J. 2013;280: 4495–4511. 10.1111/febs.12411
    1. Wegiel J, Gong CX, Hwang YW. The role of DYRK1A in neurodegenerative diseases. FEBS J. 2011;278: 236–245. 10.1111/j.1742-4658.2010.07955.x
    1. Tejedor FJ, Hämmerle B. MNB/DYRK1A as a multiple regulator of neuronal development. FEBS J. 2011;278: 223–235. 10.1111/j.1742-4658.2010.07954.x
    1. Ionescu A, Dufrasne F, Gelbcke M, Jabin I, Kiss R, Lamoral-Theys D. DYRK1A kinase inhibitors with emphasis on cancer. Mini Rev Med Chem. 2012;12: 1315–1329.
    1. Smith B, Medda F, Gokhale V, Dunckley T, Hulme C. Recent advances in the design, synthesis, and biological evaluation of selective DYRK1A inhibitors: a new avenue for a disease modifying treatment of Alzheimer's? ACS Chem Neurosci. 2012;3: 857–872. 10.1021/cn300094k
    1. Becker W, Soppa U, Tejedor FJ. DYRK1A: a potential drug target for multiple Down syndrome neuropathologies. CNS Neurol Disord Drug Targets. 2014;13: 26–33.
    1. Dierssen M. Down syndrome: the brain in trisomic mode. Nat Rev Neurosci. 2012;13: 844–858. 10.1038/nrn3314
    1. Park J, Chung KC. New perspectives of Dyrk1A role in neurogenesis and neuropathologic features of Down syndrome. Exp Neurobiol. 2013;22: 244–248. 10.5607/en.2013.22.4.244
    1. Soppa U, Schumacher J, Florencio Ortiz V, Pasqualon T, Tejedor FJ, Becker W. The Down syndrome-related protein kinase DYRK1A phosphorylates p27(Kip1) and Cyclin D1 and induces cell cycle exit and neuronal differentiation. Cell Cycle. 2014;13: 2084–2100. 10.4161/cc.29104
    1. García-Cerro S, Martínez P, Vidal V, Corrales A, Flórez J, Vidal R, et al. Overexpression of Dyrk1A is implicated in several cognitive, electrophysiological and neuromorphological alterations found in a mouse model of Down syndrome. PLOS ONE 2014;9: e106572 10.1371/journal.pone.0106572
    1. Ortiz-Abalia J, Sahún I, Altafaj X, Andreu N, Estivill X, Dierssen M, et al. Targeting Dyrk1A with AAVshRNA attenuates motor alterations in TgDyrk1A, a mouse model of Down syndrome. Am J Hum Genet. 2008;83: 479–488. 10.1016/j.ajhg.2008.09.010
    1. De la Torre R, De Sola S, Pons M, Duchon A, de Lagran MM, Farré M, et al. Epigallocatechin-3-gallate, a DYRK1A inhibitor, rescues cognitive deficits in Down syndrome mouse models and in humans. Mol Nutr Food Res. 2014;58: 278–288. 10.1002/mnfr.201300325
    1. Altafaj X, Martín ED, Ortiz-Abalia J, Valderrama A, Lao-Peregrín C, Dierssen M, et al. Normalization of Dyrk1A expression by AAV2/1-shDyrk1A attenuates hippocampal-dependent defects in the Ts65Dn mouse model of Down syndrome. Neurobiol Dis. 2013;52: 117–127. 10.1016/j.nbd.2012.11.017
    1. Aranda S, Laguna A, de la Luna S. DYRK family of protein kinases: evolutionary relationships, biochemical properties, and functional roles. FASEB J. 2011;25: 449–462. 10.1096/fj.10-165837
    1. Becker W. Emerging role of DYRK family protein kinases as regulators of protein stability in cell cycle control. Cell Cycle 2012;11: 3389–3394. 10.4161/cc.21404
    1. Di Vona C, Bezdan D, Islam AB, Salichs E, López-Bigas N, Ossowski S, et al. Chromatin-wide profiling of DYRK1A reveals a role as a gene-specific RNA polymerase II CTD kinase. Mol Cell 2015; 57: 506–520. 10.1016/j.molcel.2014.12.026
    1. Friedman E. Mirk/Dyrk1B in cancer. J Cell Biochem. 2007;102: 274–279.
    1. Ashford AL, Oxley D, Kettle J, Hudson K, Guichard S, Cook SJ, et al. A novel DYRK1B inhibitor AZ191 demonstrates that DYRK1B acts independently of GSK3β to phosphorylate cyclin D1 at Thr(286), not Thr(288). Biochem J. 2014;457: 43–56. 10.1042/BJ20130461
    1. Keramati AR, Fathzadeh M, Go GW, Singh R, Choi M, Faramarzi S, et al. A form of the metabolic syndrome associated with mutations in DYRK1B. N Engl J Med. 2014;370: 1909–1919. 10.1056/NEJMoa1301824
    1. Göckler N, Jofre G, Papadopoulos C, Soppa U, Tejedor FJ, Becker W. Harmine specifically inhibits protein kinase DYRK1A and interferes with neurite formation. FEBS J. 2009;276: 6324–6337. 10.1111/j.1742-4658.2009.07346.x
    1. Adayev T, Wegiel J, Hwang YW. Harmine is an ATP-competitive inhibitor for dual-specificity tyrosine phosphorylation-regulated kinase 1A (Dyrk1A). Arch Biochem Biophys. 2011;507: 212–218. 10.1016/j.abb.2010.12.024
    1. Ogawa Y, Nonaka Y, Goto T, Ohnishi E, Hiramatsu T, Kii I. et al. Development of a novel selective inhibitor of the Down syndrome-related kinase Dyrk1A. Nat Commun. 2010;1: 86 10.1038/ncomms1090
    1. Tahtouh T, Elkins JM, Filippakopoulos P, Soundararajan M, Burgy G, Durieu E, et al. Selectivity, cocrystal structures, and neuroprotective properties of leucettines, a family of protein kinase inhibitors derived from the marine sponge alkaloid leucettamine B. J. Med. Chem. 2012;55: 9312–9330. 10.1021/jm301034u
    1. Ginovart N, Meyer JH, Boovariwala A, Hussey D, Rabiner EA, Houle S, et al. Positron emission tomography quantification of [11C]-harmine binding to monoamine oxidase-A in the human brain. J Cereb Blood Flow Metab. 2006;26: 330–344.
    1. Hastie CJ, McLauchlan HJ, Cohen P. Assay of protein kinases using radiolabeled ATP: a protocol. Nat Protoc. 2006;1: 968–971.
    1. Drung B, Scholz C, Barbosa VA, Nazari A, Sarragiotto MH, Schmidt B. Computational & experimental evaluation of the structure/activity relationship of β-carbolines as DYRK1A inhibitors. Bioorg Med Chem Lett. 2014;24: 4854–4860. 10.1016/j.bmcl.2014.08.054
    1. Coutadeur S, Benyamine H, Delalonde L, de Oliveira C, Leblond B, Foucourt A, et al. A novel DYRK1A (Dual specificity tyrosine phosphorylation-regulated kinase 1A) inhibitor for the treatment of Alzheimer's disease: effect on Tau and amyloid pathologies in vitro. J. Neurochem. 2015;133: 440–451. 10.1111/jnc.13018
    1. Falke H, Chaikuad A, Becker A, Loaëc N, Lozach O, Abu Jhaisha S, et al. 10-Iodo-11H-indolo[3,2-c]quinoline-6-carboxylic acids are selective inhibitors of DYRK1A. J Med Chem. 2015;58:3131–3143. 10.1021/jm501994d
    1. Schmitt C, Miralinaghi P, Mariano M, Hartmann RW, Engel M. Hydroxybenzothiophene ketones are efficient pre-mRNA splicing modulators due to dual inhibition of Dyrk1A and Clk1/4. ACS Med. Chem. Lett. 2014;5: 963–7. 10.1021/ml500059y
    1. Cuny GD, Ulyanova NP, Patnaik D, Liu JF, Lin X, Auerbach K, et al. Structure-activity relationship study of beta-carboline derivatives as haspin kinase inhibitors. Bioorg Med Chem Lett. 2012;22: 2015–9. 10.1016/j.bmcl.2012.01.028
    1. Cuny GD, Robin M, Ulyanova NP, Patnaik D, Pique V, Casano G, et al. Structure-activity relationship study of acridine analogs as haspin and DYRK2 kinase inhibitors. Bioorg Med Chem Lett. 2010;20: 3491–3494.
    1. Eswaran J, Knapp S. Insights into protein kinase regulation and inhibition by large scale structural comparison. Biochim Biophys Acta. 2010;1804: 429–432. 10.1016/j.bbapap.2009.10.013
    1. Gao Y, Davies SP, Augustin M, Woodward A, Patel UA, Kovelman R, et al. A broad activity screen in support of a chemogenomic map for kinase signalling research and drug discovery. Biochem J. 2013;451, 313–28. 10.1042/BJ20121418
    1. Dai J, Sultan S, Taylor SS, Higgins JM. The kinase haspin is required for mitotic histone H3 Thr3 phosphorylation and normal metaphase chromosome alignment. Genes Dev. 2005;19: 472–488.
    1. Higgins JM. Haspin: a newly discovered regulator of mitotic chromosome behavior. Chromosoma 2010;119: 137–147. 10.1007/s00412-009-0250-4
    1. Graczyk PP. Gini coefficient: a new way to express selectivity of kinase inhibitors against a family of kinases. J Med Chem. 2007;50: 5773–5779.
    1. Cozza G, Sarno S, Ruzzene M, Girardi C, Orzeszko A, Kazimierczuk Z, et al. Exploiting the repertoire of CK2 inhibitors to target DYRK and PIM kinases. Biochim Biophys Acta 2013;1834: 1402–1409. 10.1016/j.bbapap.2013.01.018
    1. Huertas D, Soler M, Moreto J, Villanueva A, Martinez A, Vidal A, et al. Antitumor activity of a small-molecule inhibitor of the histone kinase Haspin. Oncogene 2012;31: 1408–1418. 10.1038/onc.2011.335
    1. Sitz JH, Baumgärtel K, Hämmerle B, Papadopoulos C, Hekerman P, Tejedor FJ, et al. The Down syndrome candidate dual-specificity tyrosine phosphorylation-regulated kinase 1A phosphorylates the neurodegeneration-related septin 4. Neuroscience 2008;157: 596–605. 10.1016/j.neuroscience.2008.09.034
    1. Smyth LA, Collins I. Measuring and interpreting the selectivity of protein kinase inhibitors. J Chem Biol. 2009;2: 131–151. 10.1007/s12154-009-0023-9
    1. Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Delivery Rev. 1997;23: 4–25.
    1. Hasegawa K, Yasuda SY, Teo JL, Nguyen C, McMillan M, Hsieh CL, et al. Wnt signaling orchestration with a small molecule DYRK inhibitor provides long-term xeno-free human pluripotent cell expansion. Stem Cells Transl Med. 2012;1: 18–28. 10.5966/sctm.2011-0033
    1. de Graaf K, Czajkowska H, Rottmann S, Packman LC, Lilischkis R, Lüscher B, et al. The protein kinase DYRK1A phosphorylates the splicing factor SF3b1/SAP155 at Thr434, a novel in vivo phosphorylation site. BMC Biochem. 2006;7: 7
    1. Himpel S, Tegge W, Frank R, Leder S, Joost HG, Becker W. Specificity determinants of substrate recognition by the protein kinase DYRK1A. J Biol Chem. 2000;275: 2431–2438.
    1. Becker W, Weber Y, Wetzel K, Eirmbter K, Tejedor FJ, Joost HG. Sequence characteristics, subcellular localization, and substrate specificity of DYRK-related kinases, a novel family of dual specificity protein kinases. J Biol Chem. 1998;273: 25893–25902.
    1. Stefos GC, Soppa U, Dierssen M, Becker W. NGF upregulates the plasminogen activation inhibitor-1 in neurons via the calcineurin/NFAT pathway and the Down syndrome-related proteins DYRK1A and RCAN1 attenuate this effect. PLOS ONE 2013;8: e67470 10.1371/journal.pone.0067470
    1. Mariano M, Schmitt C, Miralinaghi P, Catto M, Hartmann RW, Carotti A, et al. First selective dual inhibitors of tau phosphorylation and Beta-amyloid aggregation, two major pathogenic mechanisms in Alzheimer's disease. ACS Chem Neurosci. 2014;5: 1198–1202. 10.1021/cn5001815
    1. Bain J, Plater L, Elliott M, Shpiro N, Hastie CJ, McLauchlan H, et al. The selectivity of protein kinase inhibitors: a further update. Biochem J. 2007;408: 297–315.
    1. Schmitt C, Kail D, Mariano M, Empting M, Weber N, Paul T, et al. Design and synthesis of a library of lead-like 2,4-bisheterocyclic substituted thiophenes as selective Dyrk/Clk inhibitors. PLOS ONE 2014;9: e87851 10.1371/journal.pone.0087851

Source: PubMed

3
Tilaa