Green tea polyphenols rescue of brain defects induced by overexpression of DYRK1A

Fayçal Guedj, Catherine Sébrié, Isabelle Rivals, Aurelie Ledru, Evelyne Paly, Jean C Bizot, Desmond Smith, Edward Rubin, Brigitte Gillet, Mariona Arbones, Jean M Delabar, Fayçal Guedj, Catherine Sébrié, Isabelle Rivals, Aurelie Ledru, Evelyne Paly, Jean C Bizot, Desmond Smith, Edward Rubin, Brigitte Gillet, Mariona Arbones, Jean M Delabar

Abstract

Individuals with partial HSA21 trisomies and mice with partial MMU16 trisomies containing an extra copy of the DYRK1A gene present various alterations in brain morphogenesis. They present also learning impairments modeling those encountered in Down syndrome. Previous MRI and histological analyses of a transgenic mice generated using a human YAC construct that contains five genes including DYRK1A reveal that DYRK1A is involved, during development, in the control of brain volume and cell density of specific brain regions. Gene dosage correction induces a rescue of the brain volume alterations. DYRK1A is also involved in the control of synaptic plasticity and memory consolidation. Increased gene dosage results in brain morphogenesis defects, low BDNF levels and mnemonic deficits in these mice. Epigallocatechin gallate (EGCG) - a member of a natural polyphenols family, found in great amount in green tea leaves - is a specific and safe DYRK1A inhibitor. We maintained control and transgenic mice overexpressing DYRK1A on two different polyphenol-based diets, from gestation to adulthood. The major features of the transgenic phenotype were rescued in these mice.

Conflict of interest statement

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

Figures

Figure 1. Phenotypic correction by genotype correction…
Figure 1. Phenotypic correction by genotype correction of DYRK1A copy number.
Wild type (WT), YACtg152F7 transgenic (TG), dyrk1a (+/−) (HT) and double transgenics (TGxHT) generated by three different crossings. A: brain DYRK1A mRNA levels determined by quantitative PCR; B: in vivo MRI assessment of total brain volume (mm3); ** for p<0.01. (Mann-Whitney-Wilcoxon test; statistical significance considered to be p<0.05); C: linear regression analysis of brain volume and DYRK1A gene dosage. (R2 = 0.977).
Figure 2. Average DYRK1A protein levels in…
Figure 2. Average DYRK1A protein levels in thalamus-hypothalamus for each genotype-treatment group.
A: western blot assessment of dyrk1a and actin levels; B: average Dyrk1a protein levels for wild type (WT, n = 6), YACtg152F7 transgenic (TG, n = 6) water-fed (H2O) and in wild type (WT, n = 3), YACtg152F7 transgenic (TG, n = 7) green tea-fed (GTP). ** for p

Figure 3. Effect of GTP treatment on…

Figure 3. Effect of GTP treatment on DYRK1A-induced brain alterations.

A: weight of total brain…

Figure 3. Effect of GTP treatment on DYRK1A-induced brain alterations.
A: weight of total brain (mg) in wild type (WT, n = 26), YACtg152F7 (TG, n = 13) water-fed (H2O) and in wild type (WT, n = 13), YACtg152F7 transgenic (TG, n = 18) green tea-fed GTP; B: in vivo MRI assessment of total brain volume (mm3) in wild type (n = 10) and YACtg152F7 transgenic (n = 10) water-fed (H2O) and in wild type (n = 9) and YACtg152F7 transgenic (n = 11) green tea-fed GTP; C: in vivo MRI assessment of hypothalamus-thalamus volume (mm3) in wild type (n = 6) and YACtg152F7 transgenic (n = 6) water-fed (H2O) and in wild type (n = 5) and YACtg152F7 transgenic (n = 7) green tea-fed (GTP). (Details of the MRI experiments in supp. data). ** for p<0.01; * for p<0.05.

Figure 4. Effect of GTP treatment on…

Figure 4. Effect of GTP treatment on short- and long-term memory.

In wild type (WT,…

Figure 4. Effect of GTP treatment on short- and long-term memory.
In wild type (WT, n = 10) and YACtg152F7 transgenic (TG, n = 10) water-fed (H2O) and in wild type (WT, n = 10) and YACtg152F7 transgenic (TG, n = 10) green tea-fed (GTP, n = 10). A: Spontaneous alternation test with two sessions of ten minutes each separated by 24 h: number of alternations/total number of possible alternations ×100 (AS1). B: Object recognition test: difference in exploration time between the new and familiar objects, in percentage of total time spent exploring the two objects; 100×(N−F/N+F). ** for p2O-fed (n = 10), WT polyphenon-fed (n = 10) and TG polyphenon-fed (n = 6) (no significant differences between the three groups).

Figure 5. Effect of GTP treatment on…

Figure 5. Effect of GTP treatment on BDNF and TRKB mRNA levels.

mRNA levels determined…

Figure 5. Effect of GTP treatment on BDNF and TRKB mRNA levels.
mRNA levels determined by quantitative PCR. A: BDNF in fetal human hippocampus (EU: normal karyotype (n = 4) and T21: trisomy 21 (n = 5); B: BDNF in adult hippocampus from wild type (WT; n = 6), YACtg152F7 transgenic (TG; n = 10), water-fed (H2O) WT (n = 8), TG (n = 3), green tea-fed (GTP); C: TRKB in adult hippocampus from wild type (WT; n = 4), YACtg152F7 transgenic (TG; n = 18), water-fed (H2O) WT (n = 4), TG (n = 4) green tea-fed (GTP). ** for p
Similar articles
Cited by
References
    1. Delabar JM, Theophile D, Rahmani Z, Chettouh Z, Blouin JL, et al. Molecular mapping of twenty-four features of Down syndrome on chromosome 21. Eur J Hum Genet. 1993;1(2):114–24. - PubMed
    1. Korenberg JR, Aaltonen J, Brahe C, Cabin D, Creau N, et al. Report and abstracts of the Sixth International Workshop on Human Chromosome 21 Mapping. Cold Spring Harbor, New York, USA. May 6–8,1996. Cytogenet Cell Genet. 1997;79(1–2):21–52. - PubMed
    1. Ronan A, Fagan K, Christie L, Conroy J, Nowak NJ, et al. Familial 4.3 Mb duplication of 21q22 sheds new light on the Down syndrome critical region. J Med Genet. 2007;Jul;44(7):448–51. - PMC - PubMed
    1. Guimera J, Casas C, Pucharcos C, Solans A, Domenech A, et al. A human homologue of Drosophila minibrain (MNB) is expressed in the neuronal regions affected in Down syndrome and maps to the critical region. Hum Mol Genet. 1996;Sep;5(9):1305–10. - PubMed
    1. Tejedor F, Zhu XR, Kaltenbach E, Ackermann A, Baumann A, et al. Minibrain: a new protein kinase family involved in postembryonic neurogenesis in Drosophila. Neuron. 1995;Feb;14(2):287–301. - PubMed
Show all 46 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 3. Effect of GTP treatment on…
Figure 3. Effect of GTP treatment on DYRK1A-induced brain alterations.
A: weight of total brain (mg) in wild type (WT, n = 26), YACtg152F7 (TG, n = 13) water-fed (H2O) and in wild type (WT, n = 13), YACtg152F7 transgenic (TG, n = 18) green tea-fed GTP; B: in vivo MRI assessment of total brain volume (mm3) in wild type (n = 10) and YACtg152F7 transgenic (n = 10) water-fed (H2O) and in wild type (n = 9) and YACtg152F7 transgenic (n = 11) green tea-fed GTP; C: in vivo MRI assessment of hypothalamus-thalamus volume (mm3) in wild type (n = 6) and YACtg152F7 transgenic (n = 6) water-fed (H2O) and in wild type (n = 5) and YACtg152F7 transgenic (n = 7) green tea-fed (GTP). (Details of the MRI experiments in supp. data). ** for p<0.01; * for p<0.05.
Figure 4. Effect of GTP treatment on…
Figure 4. Effect of GTP treatment on short- and long-term memory.
In wild type (WT, n = 10) and YACtg152F7 transgenic (TG, n = 10) water-fed (H2O) and in wild type (WT, n = 10) and YACtg152F7 transgenic (TG, n = 10) green tea-fed (GTP, n = 10). A: Spontaneous alternation test with two sessions of ten minutes each separated by 24 h: number of alternations/total number of possible alternations ×100 (AS1). B: Object recognition test: difference in exploration time between the new and familiar objects, in percentage of total time spent exploring the two objects; 100×(N−F/N+F). ** for p2O-fed (n = 10), WT polyphenon-fed (n = 10) and TG polyphenon-fed (n = 6) (no significant differences between the three groups).
Figure 5. Effect of GTP treatment on…
Figure 5. Effect of GTP treatment on BDNF and TRKB mRNA levels.
mRNA levels determined by quantitative PCR. A: BDNF in fetal human hippocampus (EU: normal karyotype (n = 4) and T21: trisomy 21 (n = 5); B: BDNF in adult hippocampus from wild type (WT; n = 6), YACtg152F7 transgenic (TG; n = 10), water-fed (H2O) WT (n = 8), TG (n = 3), green tea-fed (GTP); C: TRKB in adult hippocampus from wild type (WT; n = 4), YACtg152F7 transgenic (TG; n = 18), water-fed (H2O) WT (n = 4), TG (n = 4) green tea-fed (GTP). ** for p

References

    1. Delabar JM, Theophile D, Rahmani Z, Chettouh Z, Blouin JL, et al. Molecular mapping of twenty-four features of Down syndrome on chromosome 21. Eur J Hum Genet. 1993;1(2):114–24.
    1. Korenberg JR, Aaltonen J, Brahe C, Cabin D, Creau N, et al. Report and abstracts of the Sixth International Workshop on Human Chromosome 21 Mapping. Cold Spring Harbor, New York, USA. May 6–8,1996. Cytogenet Cell Genet. 1997;79(1–2):21–52.
    1. Ronan A, Fagan K, Christie L, Conroy J, Nowak NJ, et al. Familial 4.3 Mb duplication of 21q22 sheds new light on the Down syndrome critical region. J Med Genet. 2007;Jul;44(7):448–51.
    1. Guimera J, Casas C, Pucharcos C, Solans A, Domenech A, et al. A human homologue of Drosophila minibrain (MNB) is expressed in the neuronal regions affected in Down syndrome and maps to the critical region. Hum Mol Genet. 1996;Sep;5(9):1305–10.
    1. Tejedor F, Zhu XR, Kaltenbach E, Ackermann A, Baumann A, et al. Minibrain: a new protein kinase family involved in postembryonic neurogenesis in Drosophila. Neuron. 1995;Feb;14(2):287–301.
    1. Lochhead PA, Sibbet G, Morrice N, Cleghon V. Activation-loop autophosphorylation is mediated by a novel transitional intermediate form of DYRKs. Cell. 2005;Jun 17;121(6):925–36.
    1. Woods YL, Rena G, Morrice N, Barthel A, Becker W, et al. The kinase DYRK1A phosphorylates the transcription factor FKHR at Ser329 in vitro, a novel in vivo phosphorylation site. Biochem J. 2001;May 1;355(Pt 3):597–607.
    1. Murakami N, Xie W, Lu RC, Chen-Hwang MC, Wieraszko A, et al. Phosphorylation of amphiphysin I by minibrain kinase/dual-specificity tyrosine phosphorylation-regulated kinase, a kinase implicated in Down syndrome. J Biol Chem. 2006;Aug 18;281(33):23712–24. Epub 2006 May 29.
    1. Woods YL, Cohen P, Becker W, Jakes R, Goedert M, et al. The kinase DYRK phosphorylates protein-synthesis initiation factor eIF2Bepsilon at Ser539 and the microtubule-associated protein tau at Thr212: potential role for DYRK as a glycogen synthase kinase 3-priming kinase. Biochem J. 2001;May 1;355(Pt 3):609–15.
    1. Dowjat WK, Adayev T, Kuchna I, Nowicki K, Palminiello S, et al. Trisomy-driven overexpression of DYRK1A kinase in the brain of subjects with Down syndrome. Neurosci Lett. 2007;Feb 8;413(1):77–81.
    1. Baxter LL, Moran TH, Richtsmeier JT, Troncoso J, Reeves RH. Discovery and genetic localization of Down syndrome cerebellar phenotypes using the Ts65Dn mouse. Hum Mol Genet. 2000;Jan 22;9(2):195–202.
    1. Aldridge K, Reeves RH, Olson LE, Richtsmeier JT. Differential effects of trisomy on brain shape and volume in related aneuploid mouse models. Am J Med Genet A. 2007;May 15;143A(10):1060–70.
    1. Reeves RH, Irving NG, Moran TH, Wohn A, Kitt C, et al. A mouse model for Down syndrome exhibits learning and behaviour deficits. Nat Genet. 1995;1995 Oct;11(2):177–84.
    1. Escorihuela RM, Vallina IF, Martínez-Cué C, Baamonde C, Dierssen M, et al. Impaired short- and long-term memory in Ts65Dn mice, a model for Down syndrome. Neurosci Lett. 1998;May 15;247(2–3):171–4.
    1. Fernandez F, Morishita W, Zuniga E, Nguyen J, Blank M, et al. Pharmacotherapy for cognitive impairment in a mouse model of Down syndrome. Nat Neurosci. 2007;Apr;10(4):411–3.
    1. Olson LE, Roper RJ, Sengstaken CL, Peterson EA, Aquino V, et al. Trisomy for the Down syndrome ‘critical region’ is necessary but not sufficient for brain phenotypes of trisomic mice. Hum Mol Genet. 2007;Apr 1;16(7):774–82.
    1. Dahmane N, Ghezala GA, Gosset P, Chamoun Z, Dufresne-Zacharia MC, et al. Transcriptional map of the 2.5-Mb CBR-ERG region of chromosome 21 involved in Down syndrome. Genomics. 1998;Feb 15;48(1):12–23.
    1. Hattori M, Fujiyama A, Taylor TD, Watanabe H, Yada T. The DNA sequence of human chromosome 21. Nature. 2000;May 18;405(6784):311–9. et al.; Chromosome 21 mapping and sequencing consortium. (2000) Erratum in: Nature 2000 Sep 7;407(6800):110.
    1. Smith DJ, Stevens ME, Sudanagunta SP, Bronson RT, Makhinson M, et al. Functional screening of 2 Mb of human chromosome 21q22.2 in transgenic mice implicates minibrain in learning defects associated with Down syndrome. Nat Genet. 1997;May;16(1):28–36.
    1. Branchi I, Bichler Z, Minghetti L, Delabar JM, Malchiodi-Albedi F, et al. Transgenic mouse in vivo library of human Down syndrome critical region 1: association between DYRK1A overexpression, brain development abnormalities, and cell cycle protein alteration. J Neuropathol Exp Neurol. 2004;May;63(5):429–40.
    1. Chabert C, Jamon M, Cherfouh A, Duquenne V, Smith DJ, et al. Functional analysis of genes implicated in Down syndrome: 1. Cognitive abilities in mice transpolygenic for Down Syndrome Chromosomal Region-1 (DCR-1). Behav Genet. 2004;Nov;34(6):559–69.
    1. Ahn KJ, Jeong HK, Choi HS, Ryoo SR, Kim YJ, et al. DYRK1A BAC transgenic mice show altered synaptic plasticity with learning and memory defects. Neurobiol Dis. 2006;Jun;22(3):463–72.
    1. Sebrié C, Chabert C, Ledru A, Guedj F, Po C, et al. Increased dosage of DYRK1A and brain volumetric alterations in a YAC model of partial trisomy 21. Anat Records (Hoboken) 2008;Mar;291(3):254–62.
    1. Fotaki V, Dierssen M, Alcantara S, Martinez S, Marti E, et al. Dyrk1A haploinsufficiency affects viability and causes developmental delay and abnormal brain morphology in mice. Mol Cell Biol. 2002;Sep;22(18):6636–47.
    1. Benavides-Piccione R, Dierssen M, Ballesteros-Yanez I, Martinez de Lagran M, Arbones ML, et al. Alterations in the phenotype of neocortical pyramidal cells in the Dyrk1A+/− mouse. Neurobiol Dis. 2005;Oct;20(1):115–122.
    1. Bain J, McLauchlan H, Elliott M, Cohen P. The specificities of protein kinase inhibitors: an update. Biochem J. 2003;2003 Apr 1;371(Pt 1):199–204.
    1. Adayev T, Chen-Hwang MC, Murakami N, Wegiel J, Hwang YW. Kinetic properties of a MNB/DYRK1A mutant suitable for the elucidation of biochemical pathways. Biochemistry. 2006;Oct 3;45(39):12011–9.
    1. Lin LC, Wang MN, Tseng TY, Sung JS, Tsai TH. Pharmacokinetics of (-)-epigallocatechin-3-gallate in conscious and freely moving rats and its brain regional distribution. J Agric Food Chem. 2007;Feb 21;55(4):1517–24. Epub 2007 Jan 27.
    1. Chu KO, Wang CC, Chu CY, Choy KW, Pang CP, et al. Uptake and distribution of catechins in fetal organs following in utero exposure in rats. Hum Reprod. 2007;Jan;22(1):280–7. Epub 2006 Sep.
    1. Choi JY, Park CS, Kim DJ, Cho MH, Jin BK, et al. Prevention of nitric oxide-mediated 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced Parkinson's disease in mice by tea phenolic epigallocatechin 3-gallate. Neurotoxicology. 2002;Sep;23(3):367–74.
    1. Yang DJ, Hwang LS, Lin JT. Effects of different steeping methods and storage on caffeine, catechins and gallic acid in bag tea infusions. J Chromatogr A. 2007;Jul 13;1156(1–2):312–20. Epub 2006 Dec 11.
    1. O'Doherty A, Ruf S, Mulligan C, Hildreth V, Errington ML, et al. An aneuploid mouse strain carrying human chromosome 21 with Down syndrome phenotypes. Science. 2005;Sep 23;309(5743):2033–7.
    1. Bozon B, Davis S, Laroche S. A requirement for the immediate early gene zif268 in reconsolidation of recognition memory after retrieval. Neuron. 2003;Nov 13;40(4):695–701.
    1. Hall J, Thomas KL, Everitt BJ. Rapid and selective induction of BDNF expression in the hippocampus during contextual learning. Nat Neurosci. 2000;Jun;3(6):533–5.
    1. Heldt SA, Stanek L, Chhatwal JP, Ressler KJ. Hippocampus-specific deletion of BDNF in adult mice impairs spatial memory and extinction of aversive memories. Mol Psychiatry. 2007;Jul;12(7):656–70.
    1. Barco A, Patterson S, Alarcon JM, Gromova P, Mata-Roig M, et al. Gene expression profiling of facilitated L-LTP in VP16-CREB mice reveals that BDNF is critical for the maintenance of LTP and its synaptic capture. Neuron. 2005;Oct 6;48(1):123–37.
    1. Van Praag H, Lucero MJ, Yeo GW, Stecker K, Heivand N, et al. Plant-derived flavanol (-)epicatechin enhances angiogenesis and retention of spatial memory in mice. J Neurosci. 2007;May 30;27(22):5869–78. Erratum in: J Neurosci. 2007 Aug 1;27(31):ii.
    1. Siarey RJ, Carlson EJ, Epstein CJ, Balbo A, Rapoport SI, et al. Increased synaptic depression in the Ts65Dn mouse, a model for mental retardation in Down syndrome. Neuropharmacology. 1999;Dec;38(12):1917–20.
    1. Siarey RJ, Villar AJ, Epstein CJ, Galdzicki Z. Abnormal synaptic plasticity in the Ts1Cje segmental trisomy 16 mouse model of Down syndrome. Neuropharmacology. 2005;Jul;49(1):122–8.
    1. Kleschevnikov AM, Belichenko PV, Villar AJ, Epstein CJ, Malenka RC, et al. Hippocampal long-term potentiation suppressed by increased inhibition in the Ts65Dn mouse, a genetic model of Down syndrome. J Neurosci. 2004;2004 Sep 15;24(37):8153–60.
    1. Xie W, Ramakrishna N, Wieraszko A, Hwang YW. Promotion of Neuronal Plasticity by (–)-Epigallocatechin-5 3-Gallate. Neurochem Res 2007
    1. Tanaka T, Saito H, Matsuki N. Inhibition of GABAA synaptic responses by brain-derived neurotrophic factor (BDNF) in rat hippocampus. J Neurosci. 1997;May 1;17(9):2959–66.
    1. Olofsdotter K, Lindvall O, Asztély F. Increased synaptic inhibition in dentate gyrus of mice with reduced levels of endogenous brain-derived neurotrophic factor. Neuroscience. 2000;101(3):531–9.
    1. Lee H, Bae JH, Lee SR. Protective effect of green tea polyphenol EGCG against neuronal damage and brain edema after unilateral cerebral ischemia in gerbils. J Neurosci Res. 2004;Sep 15;77(6):892–900.
    1. Chyu KY, Babbidge SM, Zhao X, Dandillaya R, Rietveld AG, et al. Differential effects of green tea-derived catechin on developing versus established atherosclerosis in apolipoprotein E-null mice. Circulation. 2004;May 25;109(20):2448–53. Epub 2004 May 10. Links.
    1. Rezai-Zadeh K, Shytle D, Sun N, Mori T, Hou H, et al. Green tea epigallocatechin-3-gallate (EGCG) modulates amyloid precursor protein cleavage and reduces cerebral amyloidosis in Alzheimer transgenic mice. J Neurosci. 2005;Sep 21;25(38):8807–14.

Source: PubMed

3
Suscribir