Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Down syndrome: the brain in trisomic mode

Key Points

  • Down syndrome results from the presence of an extra copy or major portion of human chromosome 21 (Homo sapiens autosome 21 (HSA21)), producing a genetic imbalance.

  • Our understanding of Down syndrome has shifted from a causative gene-based view to one in which genes, deregulation of non-coding elements and epigenetic factors influence the disease phenotype.

  • In Down syndrome, the ability to keep incoming information online, the performance of mental computations on such information and the storage of this information for future use are disrupted.

  • The size of certain brain regions affected in Down syndrome is correlated with performance in tests of intelligence and language.

  • HSA21-encoded proteins with master regulator functions, such as transcription or splicing efficiency of specific mRNA, may exert a combinatorial effect by promoting or inhibiting the transcription or splicing of their targets, thus spreading the effect of trisomy 21 to genes outside HSA21.

  • Many strategies have been used to model Down syndrome in mice. Mouse trisomies allow analysis of Down syndrome neurobiology, the importance of specific chromosomal regions and understanding the efficacy of treatments. Single-gene transgenesis is a complementary approach in which we may better dissect the gene-specific effects of recapitulated Down syndrome phenotypes.

  • In the past few years, we have made notable advances in finding a 'cure' for Down syndrome-linked intellectual disability based on symptomatic alleviation and individual gene function rescue.

Abstract

Down syndrome is the most common form of intellectual disability and results from one of the most complex genetic perturbations that is compatible with survival, trisomy 21. The study of brain dysfunction in this disorder has largely been based on a gene discovery approach, but we are now moving into an era of functional genome exploration, in which the effects of individual genes are being studied alongside the effects of deregulated non-coding genetic elements and epigenetic influences. Also, new data from functional neuroimaging studies are challenging our views of the cognitive phenotypes associated with Down syndrome and their pathophysiological correlates. These advances hold promise for the development of treatments for intellectual disability.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Excitation–inhibition imbalance in the hippocampal trisynaptic circuits of the Ts65Dn mouse model of Down syndrome.
Figure 2: DYRK1A in normal brain development and Down syndrome.

Similar content being viewed by others

References

  1. Megarbane, A. et al. The 50th anniversary of the discovery of trisomy 21: the past, present, and future of research and treatment of Down syndrome. Genet. Med. 11, 611–616 (2009).

    Article  PubMed  Google Scholar 

  2. Lott, I. T. & Dierssen, M. Cognitive deficits and associated neurological complications in individuals with Down's syndrome. Lancet Neurol. 9, 623–633 (2010).

    Article  PubMed  Google Scholar 

  3. Khoshnood, B., Greenlees, R., Loane, M. & Dolk, H. Paper 2: EUROCAT public health indicators for congenital anomalies in Europe. Birth Defects Res. A Clin. Mol. Teratol. 91, S16–S22 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Parker, S. E. et al. Updated National Birth Prevalence estimates for selected birth defects in the United States, 2004–2006. Birth Defects Res. A Clin. Mol. Teratol. 88, 1008–1016 (2010).

    Article  CAS  PubMed  Google Scholar 

  5. Hattori, M. et al. The DNA sequence of human chromosome 21. Nature 405, 311–319 (2000). A study that revealed the sequence and gene catalogue of the long arm of chromosome 21, which were crucial findings for Down syndrome research.

    Article  CAS  PubMed  Google Scholar 

  6. Lott, I. T. Neurological phenotypes for Down syndrome across the life span. Prog. Brain Res. 197, 101–121 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Vicari, S., Bellucci, S. & Carlesimo, G. A. Visual and spatial long-term memory: differential pattern of impairments in Williams and Down syndromes. Dev. Med. Child Neurol. 47, 305–311 (2005). A paper detailing the differences in the cognitive profiles in Down and Williams syndromes, which have helped to gain insight into the hippocampal function and the structure–function relationship in intellectual disability.

    Article  PubMed  Google Scholar 

  8. Conners, F. A., Moore, M. S., Loveall, S. J. & Merrill, E. C. Memory profiles of Down, Williams, and fragile X syndromes: implications for reading development. J. Dev. Behav. Pediatr. 32, 405–417 (2011).

    Article  PubMed  Google Scholar 

  9. Edgin, J. O., Pennington, B. F. & Mervis, C. B. Neuropsychological components of intellectual disability: the contributions of immediate, working, and associative memory. J. Intellect. Disabil. Res. 54, 406–417 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Baddeley, A. & Jarrold, C. Working memory and Down syndrome. J. Intellect. Disabil. Res. 51, 925–931 (2007). Baddeley proposed that working memory could be divided into three subsystems: one that is concerned with verbal and acoustic information (the phonological loop); a second that is concerned with the visuospatial sketchpad; and a third that is an attentionally limited control system (the central executive), on which the first and second are dependent. A fourth subsystem, the episodic buffer, has now been added to this operational model.

    Article  CAS  PubMed  Google Scholar 

  11. Lanfranchi, S., Baddeley, A., Gathercole, S. & Vianello, R. Working memory in Down syndrome: is there a dual task deficit? J. Intellect. Disabil. Res. 56, 157–166 (2011).

    Article  PubMed  Google Scholar 

  12. Dierssen, M., Herault, Y. & Estivill, X. Aneuploidy: from a physiological mechanism of variance to Down syndrome. Physiol. Rev. 89, 887–920 (2009).

    Article  CAS  PubMed  Google Scholar 

  13. Chapman, R. S. & Hesketh, L. J. Behavioral phenotype of individuals with Down syndrome. Ment. Retard. Dev. Disabil. Res. Rev. 6, 84–95 (2000).

    Article  CAS  PubMed  Google Scholar 

  14. Laws, G. & Bishop, D. V. A comparison of language abilities in adolescents with Down syndrome and children with specific language impairment. J. Speech Lang. Hear. Res. 46, 1324–1339 (2003).

    Article  PubMed  Google Scholar 

  15. Purser, H. R. M. & Jarrold, C. Impaired verbal short-term memory in Down syndrome reflects a capacity limitation rather than atypically rapid forgetting. J. Exp. Child Psychol. 91, 1–23 (2005).

    Article  Google Scholar 

  16. Zimmer, H. D. Visual and spatial working memory: from boxes to networks. Neurosci. Biobehav. Rev. 32, 1373–1395 (2008).

    Article  PubMed  Google Scholar 

  17. Visu-Petra, L., Benga, O., Tincas, I. & Miclea, M. Visual-spatial processing in children and adolescents with Down's syndrome: a computerized assessment of memory skills. J. Intellect. Disabil. Res. 51, 942–952 (2007).

    Article  CAS  PubMed  Google Scholar 

  18. Lanfranchi, S., Carretti, B., Spano, G. & Cornoldi, C. A specific deficit in visuospatial simultaneous working memory in Down syndrome. J. Intellect. Disabil. Res. 53, 474–483 (2009).

    Article  CAS  PubMed  Google Scholar 

  19. Colom, R., Jung, R. E. & Haier, R. J. General intelligence and memory span: evidence for a common neuroanatomic framework. Cogn. Neuropsychol. 24, 867–878 (2007).

    Article  PubMed  Google Scholar 

  20. Jarrold, C. & Towse, J. N. Individual differences in working memory. Neuroscience 139, 39–50 (2006).

    Article  CAS  PubMed  Google Scholar 

  21. Nairne, J. S. Remembering over the short-term: the case against the standard model. Annu. Rev. Psychol. 53, 53–81 (2002).

    Article  PubMed  Google Scholar 

  22. Nash, H. & Heath, J. The role of vocabulary, working memory and inference making ability in reading comprehension in Down syndrome. Res. Dev. Disabil. 32, 1782–1791 (2011).

    Article  PubMed  Google Scholar 

  23. Saito, S., Jarrold, C. & Riby, D. M. Exploring the forgetting mechanisms in working memory: evidence from a reasoning span test. Q. J. Exp. Psychol. 62, 1401–1419 (2009).

    Article  Google Scholar 

  24. Lanfranchi, S., Jerman, O., Dal Pont, E., Alberti, A. & Vianello, R. Executive function in adolescents with Down Syndrome. J. Intellect. Disabil. Res. 54, 308–319 (2010).

    Article  CAS  PubMed  Google Scholar 

  25. Lemons, C. J. & Fuchs, D. Phonological awareness of children with Down syndrome: its role in learning to read and the effectiveness of related interventions. Res. Dev. Disabil. 31, 316–330 (2010).

    Article  PubMed  Google Scholar 

  26. Jarrold, C., Cocksey, J. & Dockerill, E. Phonological similarity and lexicality effects in children's verbal short-term memory: concerns about the interpretation of probed recall data. Q. J. Exp. Psychol. 61, 324–340 (2008).

    Article  Google Scholar 

  27. Fidler, D. J., Most, D. E. & Guiberson, M. M. Neuropsychological correlates of word identification in Down syndrome. Res. Dev. Disabil. 26, 487–501 (2005).

    Article  PubMed  Google Scholar 

  28. Jarrold, C., Baddeley, A. D. & Phillips, C. E. Verbal short-term memory in Down syndrome: a problem of memory, audition, or speech? J. Speech Lang. Hear. Res. 45, 531–544 (2002).

    Article  PubMed  Google Scholar 

  29. Brock, J. & Jarrold, C. Serial order reconstruction in Down syndrome: evidence for a selective deficit in verbal short-term memory. J. Child Psychol. Psychiatry 46, 304–316 (2005).

    Article  PubMed  Google Scholar 

  30. Robertson Ringenbach, S. D., Chua, R., Maraj, B. K., Kao, J. C. & Weeks, D. J. Bimanual coordination dynamics in adults with Down syndrome. Motor Control 6, 388–407 (2002).

    Article  PubMed  Google Scholar 

  31. Robertson, S. D., Van Gemmert, A. W. & Maraj, B. K. Auditory information is beneficial for adults with Down syndrome in a continuous bimanual task. Acta Psychol. 110, 213–229 (2002).

    Article  Google Scholar 

  32. Elliott, D., Weeks, D. J. & Gray, S. Manual and oral praxis in adults with Down's syndrome. Neuropsychologia 28, 1307–1315 (1990).

    Article  CAS  PubMed  Google Scholar 

  33. Elliott, D. & Weeks, D. J. Cerebral specialization and the control of oral and limb movements for individuals with Down's syndrome. J. Mot Behav. 22, 6–18 (1990).

    Article  CAS  PubMed  Google Scholar 

  34. Welsh, T. N. & Elliott, D. Gender differences in a dichotic listening and movement task: lateralization or strategy? Neuropsychologia 39, 25–35 (2001).

    Article  CAS  PubMed  Google Scholar 

  35. Brunamonti, E. et al. Cognitive control of movement in Down syndrome. Res. Dev. Disabil. 32, 1792–1797 (2011).

    Article  PubMed  Google Scholar 

  36. Davis, W. E. & Kelso, J. A. Analysis of “invariant characteristics” in the motor control of down's syndrome and normal subjects. J. Mot. Behav. 14, 194–212 (1982).

    Article  CAS  PubMed  Google Scholar 

  37. Virji-Babul, N. et al. Altered brain dynamics during voluntary movement in individuals with Down syndrome. Neuroreport 22, 358–364 (2011).

    Article  PubMed  Google Scholar 

  38. Vicari, S., Bellucci, S. & Carlesimo, G. A. Implicit and explicit memory: a functional dissociation in persons with Down syndrome. Neuropsychologia 38, 240–251 (2000).

    Article  CAS  PubMed  Google Scholar 

  39. Vakil, E. & Lifshitz-Zehavi, H. Solving the Raven Progressive Matrices by adults with intellectual disability with/without Down syndrome: different cognitive patterns as indicated by eye-movements. Res. Dev. Disabil. 33, 645–654 (2012).

    Article  PubMed  Google Scholar 

  40. Pinter, J. D., Eliez, S., Schmitt, J. E., Capone, G. T. & Reiss, A. L. Neuroanatomy of Down's syndrome: a high-resolution MRI study. Am. J. Psychiatry 158, 1659–1665 (2001). A seminal paper showing the altered neuroanatomy in patients with Down syndrome patients using MRI.

    Article  CAS  PubMed  Google Scholar 

  41. Raz, N. et al. Selective neuroanatomic abnormalities in Down's syndrome and their cognitive correlates: evidence from MRI morphometry. Neurology 45, 356–366 (1995).

    Article  CAS  PubMed  Google Scholar 

  42. Menghini, D., Costanzo, F. & Vicari, S. Relationship between brain and cognitive processes in Down syndrome. Behav. Genet. 41, 381–393 (2011). This was one of several studies that attempted to correlate structural abnormalities with cognitive efficiency.

    Article  PubMed  Google Scholar 

  43. Jernigan, T. L., Bellugi, U., Sowell, E., Doherty, S. & Hesselink, J. R. Cerebral morphologic distinctions between Williams and Down syndromes. Arch. Neurol. 50, 186–191 (1993).

    Article  CAS  PubMed  Google Scholar 

  44. Jarrold, C., Baddeley, A. D. & Phillips, C. Long-term memory for verbal and visual information in Down syndrome and Williams syndrome: performance on the Doors and People test. Cortex 43, 233–247 (2007).

    Article  PubMed  Google Scholar 

  45. Vicari, S., Bellucci, S. & Carlesimo, G. A. Evidence from two genetic syndromes for the independence of spatial and visual working memory. Dev. Med. Child Neurol. 48, 126–131 (2006).

    Article  PubMed  Google Scholar 

  46. Di Filippo, M. et al. Impaired plasticity at specific subset of striatal synapses in the Ts65Dn mouse model of Down syndrome. Biol. Psychiatry 67, 666–671 (2010).

    Article  CAS  PubMed  Google Scholar 

  47. Tomporowski, P. D., Hayden, A. M. & Applegate, B. Effects of background event rate on sustained attention of mentally retarded and nonretarded adults. Am. J. Ment. Retard. 94, 499–508 (1990).

    CAS  PubMed  Google Scholar 

  48. Rigoldi, C. et al. Gait analysis and cerebral volumes in Down's syndrome. Funct. Neurol. 24, 147–152 (2009).

    CAS  PubMed  Google Scholar 

  49. Jacola, L. M. et al. Functional magnetic resonance imaging of cognitive processing in young adults with Down syndrome. Am. J. Intellect. Dev. Disabil. 116, 344–359 (2011).

    Article  PubMed  Google Scholar 

  50. Colom, R., Karama, S., Jung, R. E. & Haier, R. J. Human intelligence and brain networks. Dialogues Clin. Neurosci. 12, 489–501 (2010).

    PubMed  PubMed Central  Google Scholar 

  51. Fabbro, F., Libera, L. & Tavano, A. A callosal transfer deficit in children with developmental language disorder. Neuropsychologia 40, 1541–1546 (2002).

    Article  PubMed  Google Scholar 

  52. Pennington, B. F., Moon, J., Edgin, J., Stedron, J. & Nadel, L. The neuropsychology of Down syndrome: evidence for hippocampal dysfunction. Child Dev. 74, 75–93 (2003). A seminal paper showing hippocampal dysfunction in patients with Down syndrome that was revealed using specific neuropsychological tests that were based on mouse work.

    Article  PubMed  Google Scholar 

  53. Sturgeon, X. & Gardiner, K. J. Transcript catalogs of human chromosome 21 and orthologous chimpanzee and mouse regions. Mamm. Genome 22, 261–271 (2011).

    Article  PubMed  Google Scholar 

  54. Delabar, J. M. et al. Molecular mapping of twenty-four features of Down syndrome on chromosome 21. Eur. J. Hum. Genet. 1, 114–124 (1993).

    Article  CAS  PubMed  Google Scholar 

  55. Korenberg, J. R. et al. Down syndrome phenotypes: the consequences of chromosomal imbalance. Proc. Natl Acad. Sci. USA 91, 4997–5001 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Korbel, J. O. et al. The genetic architecture of Down syndrome phenotypes revealed by high-resolution analysis of human segmental trisomies. Proc. Natl Acad. Sci. USA 106, 12031–12036 (2009). This study shows that different regions of HSA21 contribute to different disease-related phenotypes, arguing against a single critical region in HSA21 (also see reference 57).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Lyle, R. et al. Genotype–phenotype correlations in Down syndrome identified by array CGH in 30 cases of partial trisomy and partial monosomy chromosome 21. Eur. J. Hum. Genet. 17, 454–466 (2009).

    Article  CAS  PubMed  Google Scholar 

  58. Dierssen, M. et al. Functional genomics of Down syndrome: a multidisciplinary approach. J. Neural Transm. Suppl. 61, 131–148 (2001).

    Google Scholar 

  59. Pritchard, M., Reeves, R. H., Dierssen, M., Patterson, D. & Gardiner, K. J. Down syndrome and the genes of human chromosome 21: current knowledge and future potentials. Report on the Expert workshop on the biology of chromosome 21 genes: towards gene-phenotype correlations in Down syndrome. Washington D.C., September 28-October 1, 2007. Cytogenet. Genome Res. 121, 67–77 (2008).

    Article  CAS  PubMed  Google Scholar 

  60. Pritchard, M. A. & Kola, I. The “gene dosage effect” hypothesis versus the “amplified developmental instability” hypothesis in Down syndrome. J. Neural Transm. Suppl. 57, 293–303 (1999).

    CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Toiber, D. et al. Engineering DYRK1A overdosage yields Down syndrome-characteristic cortical splicing aberrations. Neurobiol. Dis. 40, 348–359 (2010). This study shows that a Down syndrome candidate gene on HSA21, DYRK1A , has master regulatory functions in transcription and mRNA splicing and thus may exert wide-ranging effects, acting on genes that are found on HSA21 and other chromosomes. Also see references 63–65.

    Article  CAS  PubMed  Google Scholar 

  63. Kahlem, P. et al. Transcript level alterations reflect gene dosage effects across multiple tissues in a mouse model of down syndrome. Genome Res. 14, 1258–1267 (2004). This study tested the hypothesis that HSA21 transcripts are overexpressed by about 50% in trisomic cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Arron, J. R. et al. NFAT dysregulation by increased dosage of DSCR1 and DYRK1A on chromosome 21. Nature 441, 595–600 (2006). DYRK1A phosphorylates NFAT while calcipressin 1 phosphorylation at Thr192 by DYRK1A enhances the ability of calcipressin 1 to inhibit the phosphatase activity of calcineurin, leading to reduced NFAT transcriptional activity.

    Article  CAS  PubMed  Google Scholar 

  65. Vilardell, M. et al. Meta-analysis of heterogeneous Down Syndrome data reveals consistent genome-wide dosage effects related to neurological processes. BMC Genomics 12, 229 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Ferrando-Miguel, R., Cheon, M. S., Yang, J. W. & Lubec, G. Overexpression of transcription factor BACH1 in fetal Down syndrome brain. J. Neural Transm. Suppl. 67, 193–205 (2003).

    Article  CAS  Google Scholar 

  67. Shim, K. S., Ferrando-Miguel, R. & Lubec, G. Aberrant protein expression of transcription factors BACH1 and ERG, both encoded on chromosome 21, in brains of patients with Down syndrome and Alzheimer's disease. J. Neural Transm. Suppl. 67, 39–49 (2003).

    Article  CAS  Google Scholar 

  68. Osato, M. & Ito, Y. Increased dosage of the RUNX1/AML1 gene: a third mode of RUNX leukemia? Crit. Rev. Eukaryot. Gene Expr. 15, 217–228 (2005).

    Article  CAS  PubMed  Google Scholar 

  69. Altafaj, X. et al. Neurodevelopmental delay, motor abnormalities and cognitive deficits in transgenic mice overexpressing Dyrk1A (minibrain), a murine model of Down's syndrome. Hum. Mol. Genet. 10, 1915–1923 (2001). Dyrk1a is a major candidate gene for Down syndrome. This paper reports the construction of the first transgenic mouse model overexpressing this kinase. Many other DYRK1A overexpression models have subsequently been created.

    Article  CAS  PubMed  Google Scholar 

  70. Ahn, K. J. et al. DYRK1A BAC transgenic mice show altered synaptic plasticity with learning and memory defects. Neurobiol. Dis. 22, 463–472 (2006).

    Article  CAS  PubMed  Google Scholar 

  71. Ferrer, I. et al. Constitutive Dyrk1A is abnormally expressed in Alzheimer disease, Down syndrome, Pick disease, and related transgenic models. Neurobiol. Dis. 20, 392–400 (2005). This was one of the first papers suggesting a role for DYRK1A in Alzheimer's disease.

    Article  CAS  PubMed  Google Scholar 

  72. Fuentes, J. J. et al. A new human gene from the Down syndrome critical region encodes a proline-rich protein highly expressed in fetal brain and heart. Hum. Mol. Genet. 4, 1935–1944 (1995).

    Article  CAS  PubMed  Google Scholar 

  73. Shapiro, B. L. Amplified developmental instability in Down's syndrome. Ann. Hum. Genet. 38, 429–437 (1975). The amplified developmental instability hypothesis, which is proposed in this paper, states that the dosage imbalance of HSA21 leads to a non-specific disturbance of cellular homeostasis.

    Article  CAS  PubMed  Google Scholar 

  74. Shapiro, B. L. Developmental instability of the cerebellum and its relevance to Down syndrome. J. Neural Transm. Suppl. 61, 11–34 (2001).

    Google Scholar 

  75. Olson, L. E. et al. Down syndrome mouse models Ts65Dn, Ts1Cje, and Ms1Cje/Ts65Dn exhibit variable severity of cerebellar phenotypes. Dev. Dyn. 230, 581–589 (2004). This paper made an important contribution to the understanding of Down syndrome cerebellar phenotypes. The cerebellar alterations in Down syndrome were thought to participate mainly in motor phenotypes, but abnormalities in this region are now thought to also affect cognitive processes.

    Article  CAS  PubMed  Google Scholar 

  76. Yu, T. et al. Effects of individual segmental trisomies of human chromosome 21 syntenic regions on hippocampal long-term potentiation and cognitive behaviors in mice. Brain Res. 1366, 162–171 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Kuhn, D. E. et al. Human chromosome 21-derived miRNAs are overexpressed in down syndrome brains and hearts. Biochem. Biophys. Res. Commun. 370, 473–477 (2008). This study showed that HSA21-derived microRNAs are overexpressed in Down syndrome brain and heart specimens, leading to improper repression of specific target proteins that are linked to specific phenotypes.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Kuhn, D. E. et al. Chromosome 21-derived microRNAs provide an etiological basis for aberrant protein expression in human Down syndrome brains. J. Biol. Chem. 285, 1529–1543 (2010).

    Article  CAS  PubMed  Google Scholar 

  79. Loudin, M. G. et al. Genomic profiling in Down syndrome acute lymphoblastic leukemia identifies histone gene deletions associated with altered methylation profiles. Leukemia 25, 1555–1563 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Pletcher, M. T., Wiltshire, T., Cabin, D. E., Villanueva, M. & Reeves, R. H. Use of comparative physical and sequence mapping to annotate mouse chromosome 16 and human chromosome 21. Genomics 74, 45–54 (2001).

    Article  CAS  PubMed  Google Scholar 

  81. Davisson, M. T. et al. Segmental trisomy as a mouse model for Down syndrome. Prog. Clin. Biol. Res. 384, 117–133 (1993). The creation of the first viable trisomic mouse (Ts65Dn) was a revolution in the field of Down syndrome research and provided the best in vivo model for Down syndrome. Also see reference 82.

    CAS  PubMed  Google Scholar 

  82. Reeves, R. H. et al. A mouse model for Down syndrome exhibits learning and behaviour deficits. Nature Genet. 11, 177–184 (1995).

    Article  CAS  PubMed  Google Scholar 

  83. Dierssen, M. et al. Alterations of neocortical pyramidal cell phenotype in the Ts65Dn mouse model of Down syndrome: effects of environmental enrichment. Cereb. Cortex 13, 758–764 (2003).

    Article  CAS  PubMed  Google Scholar 

  84. Sago, H. et al. Ts1Cje, a partial trisomy 16 mouse model for Down syndrome, exhibits learning and behavioral abnormalities. Proc. Natl Acad. Sci. USA 95, 6256–6261 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Villar, A. J. et al. Identification and characterization of a new Down syndrome model, Ts[Rb(12.1716)]2Cje, resulting from a spontaneous Robertsonian fusion between T(171)65Dn and mouse chromosome 12. Mamm. Genome 16, 79–90 (2005).

    Article  CAS  PubMed  Google Scholar 

  86. Ishihara, K. et al. Enlarged brain ventricles and impaired neurogenesis in the Ts1Cje and Ts2Cje mouse models of Down syndrome. Cereb. Cortex 20, 1131–1143 (2010).

    Article  PubMed  Google Scholar 

  87. Olson, L. E. et al. Trisomy for the Down syndrome 'critical region' is necessary but not sufficient for brain phenotypes of trisomic mice. Hum. Mol. Genet. 16, 774–782 (2007). This study explored the role of the DSCR and presented evidence that questioned the DSCR hypothesis.

    Article  CAS  PubMed  Google Scholar 

  88. Belichenko, N. P. et al. The “Down syndrome critical region” is sufficient in the mouse model to confer behavioral, neurophysiological, and synaptic phenotypes characteristic of Down syndrome. J. Neurosci. 29, 5938–5948 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Olson, L. E., Richtsmeier, J. T., Leszl, J. & Reeves, R. H. A chromosome 21 critical region does not cause specific Down syndrome phenotypes. Science 306, 687–690 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Aldridge, K., Reeves, R. H., Olson, L. E. & Richtsmeier, J. T. Differential effects of trisomy on brain shape and volume in related aneuploid mouse models. Am. J. Med. Genet. A 143A, 1060–1070 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  91. Duchon, A. et al. The telomeric part of the human chromosome 21 from Cstb to Prmt2 is not necessary for the locomotor and short-term memory deficits observed in the Tc1 mouse model of Down syndrome. Behav. Brain Res. 217, 271–281 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. O'Doherty, A. et al. An aneuploid mouse strain carrying human chromosome 21 with Down syndrome phenotypes. Science 309, 2033–2037 (2005). This paper details the creation of the first 'humanized' Down syndrome mouse model.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Morice, E. et al. Preservation of long-term memory and synaptic plasticity despite short-term impairments in the Tc1 mouse model of Down syndrome. Learn. Mem. 15, 492–500 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Galante, M. et al. Impairments in motor coordination without major changes in cerebellar plasticity in the Tc1 mouse model of Down syndrome. Hum. Mol. Genet. 18, 1449–1463 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Reynolds, T. The triple test as a screening technique for Down syndrome: reliability and relevance. Int. J. Womens Health 2, 83–88 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Li, Z. et al. Duplication of the entire 22.9 Mb human chromosome 21 syntenic region on mouse chromosome 16 causes cardiovascular and gastrointestinal abnormalities. Hum. Mol. Genet. 16, 1359–1366 (2007).

    Article  CAS  PubMed  Google Scholar 

  97. Belichenko, P. V., Kleschevnikov, A. M., Salehi, A., Epstein, C. J. & Mobley, W. C. Synaptic and cognitive abnormalities in mouse models of Down syndrome: exploring genotype-phenotype relationships. J. Comp. Neurol. 504, 329–345 (2007).

    Article  CAS  PubMed  Google Scholar 

  98. Escorihuela, R. M. et al. A behavioral assessment of Ts65Dn mice: a putative Down syndrome model. Neurosci. Lett. 199, 143–146 (1995).

    Article  CAS  PubMed  Google Scholar 

  99. Nambu, J. R., Lewis, J. O., Wharton, K. A. Jr & Crews, S. T. The Drosophila single-minded gene encodes a helix-loop-helix protein that acts as a master regulator of CNS midline development. Cell 67, 1157–1167 (1991).

    Article  CAS  PubMed  Google Scholar 

  100. Rachidi, M. et al. Spatial and temporal localization during embryonic and fetal human development of the transcription factor SIM2 in brain regions altered in Down syndrome. Int. J. Dev. Neurosci. 23, 475–484 (2005).

    Article  CAS  PubMed  Google Scholar 

  101. Chrast, R. et al. Mice trisomic for a bacterial artificial chromosome with the single-minded 2 gene (Sim2) show phenotypes similar to some of those present in the partial trisomy 16 mouse models of Down syndrome. Hum. Mol. Genet. 9, 1853–1864 (2000).

    Article  CAS  PubMed  Google Scholar 

  102. Chrast, R. et al. The mouse brain transcriptome by SAGE: differences in gene expression between P30 brains of the partial trisomy 16 mouse model of Down syndrome (Ts65Dn) and normals. Genome Res. 10, 2006–2021 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Harashima, C. et al. Elevated expression of the G-protein-activated inwardly rectifying potassium channel 2 (GIRK2) in cerebellar unipolar brush cells of a Down syndrome mouse model. Cell. Mol. Neurobiol. 26, 719–734 (2006).

    Article  CAS  PubMed  Google Scholar 

  104. Cooper, A. et al. Trisomy of the G protein-coupled K+ channel gene, Kcnj6, affects reward mechanisms, cognitive functions, and synaptic plasticity in mice. Proc. Natl Acad. Sci. USA 109, 2642–2647 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  105. Alves-Sampaio, A., Troca-Marin, J. A. & Montesinos, M. L. NMDA-mediated regulation of DSCAM dendritic local translation is lost in a mouse model of Down's syndrome. J. Neurosci. 30, 13537–13548 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Gotti, S., Caricati, E. & Panzica, G. Alterations of brain circuits in Down syndrome murine models. J. Chem. Neuroanat. 42, 317–326 (2011).

    Article  CAS  PubMed  Google Scholar 

  107. Liu, C. et al. Mouse models for down syndrome-associated developmental cognitive disabilities. Dev. Neurosci. 33, 404–413 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Dierssen, M. et al. Murine models for Down syndrome. Physiol. Behav. 73, 859–871 (2001).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Smith, D. J., Zhu, Y., Zhang, J., Cheng, J. F. & Rubin, E. M. Construction of a panel of transgenic mice containing a contiguous 2-Mb set of YAC/P1 clones from human chromosome 21q22.2. Genomics 27, 425–434 (1995). Smith et al . created the first in vivo library of mice bearing contiguous partial trisomies.

    Article  CAS  PubMed  Google Scholar 

  111. Guedj, F. et al. DYRK1A: a master regulatory protein controlling brain growth. Neurobiol. Dis. 46, 190–203 (2012).

    Article  CAS  PubMed  Google Scholar 

  112. Smith, D. J. & Rubin, E. M. Functional screening and complex traits: human 21q22.2 sequences affecting learning in mice. Hum. Mol. Genet. 6, 1729–1733 (1997).

    Article  CAS  PubMed  Google Scholar 

  113. Kleschevnikov, A. M. et al. Hippocampal long-term potentiation suppressed by increased inhibition in the Ts65Dn mouse, a genetic model of Down syndrome. J. Neurosci. 24, 8153–8160 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Yuste, R. Dendritic spines and distributed circuits. Neuron 71, 772–781 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Dierssen, M. & Ramakers, G. J. Dendritic pathology in mental retardation: from molecular genetics to neurobiology. Genes Brain Behav. 5, 48–60 (2006).

    Article  CAS  PubMed  Google Scholar 

  116. Galdzicki, Z. & Siarey, R. J. Understanding mental retardation in Down's syndrome using trisomy 16 mouse models. Genes Brain Behav. 2, 167–178 (2003).

    Article  CAS  PubMed  Google Scholar 

  117. Siarey, R. J., Stoll, J., Rapoport, S. I. & Galdzicki, Z. Altered long-term potentiation in the young and old Ts65Dn mouse, a model for Down Syndrome. Neuropharmacology 36, 1549–1554 (1997).

    Article  CAS  PubMed  Google Scholar 

  118. Kurt, M. A., Davies, D. C., Kidd, M., Dierssen, M. & Florez, J. Synaptic deficit in the temporal cortex of partial trisomy 16 (Ts65Dn) mice. Brain Res. 858, 191–197 (2000). This is the first paper describing the excitation–inhibition imbalance in the hippocampus of a Down syndrome trisomic mouse model (Ts65Dn).

    Article  CAS  PubMed  Google Scholar 

  119. Pereira, P. L. et al. A new mouse model for the trisomy of the Abcg1−U2af1 region reveals the complexity of the combinatorial genetic code of down syndrome. Hum. Mol. Genet. 18, 4756–4769 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Martinez de Lagran, M. et al. Dyrk1A influences neuronal morphogenesis through regulation of cytoskeletal dynamics in mammalian cortical neurons. Cereb. Cortex 2 Jan 2012 (doi:10.1093/cercor/bhr362).

  121. Kurt, M. A., Kafa, M. I., Dierssen, M. & Davies, D. C. Deficits of neuronal density in CA1 and synaptic density in the dentate gyrus, CA3 and CA1, in a mouse model of Down syndrome. Brain Res. 1022, 101–109 (2004).

    Article  CAS  PubMed  Google Scholar 

  122. Belichenko, P. V. et al. Excitatory-inhibitory relationship in the fascia dentata in the Ts65Dn mouse model of Down syndrome. J. Comp. Neurol. 512, 453–466 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  123. Siarey, R. J. et al. Increased synaptic depression in the Ts65Dn mouse, a model for mental retardation in Down syndrome. Neuropharmacology 38, 1917–1920 (1999).

    Article  CAS  PubMed  Google Scholar 

  124. Popov, V. I., Kleschevnikov, A. M., Klimenko, O. A., Stewart, M. G. & Belichenko, P. V. Three-dimensional synaptic ultrastructure in the dentate gyrus and hippocampal area CA3 in the Ts65Dn mouse model of Down syndrome. J. Comp. Neurol. 519, 1338–1354 (2011).

    Article  CAS  PubMed  Google Scholar 

  125. Escorihuela, R. M. et al. Early environmental stimulation produces long-lasting changes on β-adrenoceptor transduction system. Neurobiol. Learn. Mem. 64, 49–57 (1995).

    Article  CAS  PubMed  Google Scholar 

  126. Coussons-Read, M. E. & Crnic, L. S. Behavioral assessment of the Ts65Dn mouse, a model for Down syndrome: altered behavior in the elevated plus maze and open field. Behav. Genet. 26, 7–13 (1996).

    Article  CAS  PubMed  Google Scholar 

  127. Hyde, L. A., Crnic, L. S., Pollock, A. & Bickford, P. C. Motor learning in Ts65Dn mice, a model for Down syndrome. Dev. Psychobiol. 38, 33–45 (2001).

    Article  CAS  PubMed  Google Scholar 

  128. Driscoll, L. L. et al. Impaired sustained attention and error-induced stereotypy in the aged Ts65Dn mouse: a mouse model of Down syndrome and Alzheimer's disease. Behav. Neurosci. 118, 1196–1205 (2004).

    Article  PubMed  Google Scholar 

  129. Escorihuela, R. M. et al. Impaired short- and long-term memory in Ts65Dn mice, a model for Down syndrome. Neurosci. Lett. 247, 171–174 (1998).

    Article  CAS  PubMed  Google Scholar 

  130. Martinez-Cue, C. et al. Differential effects of environmental enrichment on behavior and learning of male and female Ts65Dn mice, a model for Down syndrome. Behav. Brain Res. 134, 185–200 (2002).

    Article  PubMed  Google Scholar 

  131. Baamonde, C., Martinez-Cue, C., Florez, J. & Dierssen, M. G-protein-associated signal transduction processes are restored after postweaning environmental enrichment in Ts65Dn, a Down Syndrome mouse model. Dev. Neurosci. 33, 442–450 (2011).

    Article  CAS  PubMed  Google Scholar 

  132. Casanova, M. F., Walker, L. C., Whitehouse, P. J. & Price, D. L. Abnormalities of the nucleus basalis in Down's syndrome. Ann. Neurol. 18, 310–313 (1985).

    Article  CAS  PubMed  Google Scholar 

  133. Mann, D. M., Yates, P. O. & Hawkes, J. The pathology of the human locus ceruleus. Clin. Neuropathol. 2, 1–7 (1983).

    CAS  PubMed  Google Scholar 

  134. Salehi, A. et al. Restoration of norepinephrine-modulated contextual memory in a mouse model of Down syndrome. Sci. Transl. Med. 1, 7ra17 (2009).

    Article  CAS  PubMed  Google Scholar 

  135. Chang, Q. & Gold, P. E. Age-related changes in memory and in acetylcholine functions in the hippocampus in the Ts65Dn mouse, a model of Down syndrome. Neurobiol. Learn. Mem. 89, 167–177 (2008).

    Article  CAS  PubMed  Google Scholar 

  136. Rueda, N., Florez, J. & Martinez-Cue, C. Effects of chronic administration of SGS-111 during adulthood and during the pre- and post-natal periods on the cognitive deficits of Ts65Dn mice, a model of Down syndrome. Behav. Brain Res. 188, 355–367 (2008).

    Article  CAS  PubMed  Google Scholar 

  137. Holtzman, D. M. et al. Developmental abnormalities and age-related neurodegeneration in a mouse model of Down syndrome. Proc. Natl Acad. Sci. USA 93, 13333–13338 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Prasher, V. P., Fung, N. & Adams, C. Rivastigmine in the treatment of dementia in Alzheimer's disease in adults with Down syndrome. Int. J. Geriatr. Psychiatry 20, 496–497 (2005).

    Article  CAS  PubMed  Google Scholar 

  139. Van der Molen, M. J. et al. Attentional set-shifting in fragile X syndrome. Brain Cogn. 78, 206–217 (2012).

    Article  CAS  PubMed  Google Scholar 

  140. Hanson, J. E., Blank, M., Valenzuela, R. A., Garner, C. C. & Madison, D. V. The functional nature of synaptic circuitry is altered in area CA3 of the hippocampus in a mouse model of Down's syndrome. J. Physiol. 579, 53–67 (2007).

    Article  CAS  PubMed  Google Scholar 

  141. Fagiolini, M. & Hensch, T. K. Inhibitory threshold for critical-period activation in primary visual cortex. Nature 404, 183–186 (2000).

    Article  CAS  PubMed  Google Scholar 

  142. Southwell, D. G., Froemke, R. C., Alvarez-Buylla, A., Stryker, M. P. & Gandhi, S. P. Cortical plasticity induced by inhibitory neuron transplantation. Science 327, 1145–1148 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Fernandez, F. et al. Pharmacotherapy for cognitive impairment in a mouse model of Down syndrome. Nature Neurosci. 10, 411–413 (2007).

    Article  CAS  PubMed  Google Scholar 

  144. Rueda, N., Florez, J. & Martinez-Cue, C. Chronic pentylenetetrazole but not donepezil treatment rescues spatial cognition in Ts65Dn mice, a model for Down syndrome. Neurosci. Lett. 433, 22–27 (2008).

    Article  CAS  PubMed  Google Scholar 

  145. Owens, D. F. & Kriegstein, A. R. Developmental neurotransmitters? Neuron 36, 989–991 (2002).

    Article  CAS  PubMed  Google Scholar 

  146. Baroncelli, L. et al. Brain plasticity and disease: a matter of inhibition. Neural Plast. 2011, 286073 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  147. Owens, D. F. & Kriegstein, A. R. Is there more to GABA than synaptic inhibition? Nature Rev. Neurosci. 3, 715–727 (2002).

    Article  CAS  Google Scholar 

  148. Becker, L., Mito, T., Takashima, S. & Onodera, K. Growth and development of the brain in Down syndrome. Prog. Clin. Biol. Res. 373, 133–152 (1991).

    CAS  PubMed  Google Scholar 

  149. Braudeau, J. et al. Specific targeting of the GABA-A receptor α5 subtype by a selective inverse agonist restores cognitive deficits in Down syndrome mice. J. Psychopharmacol. 25, 1030–1042 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Costa, A. C., Scott-McKean, J. J. & Stasko, M. R. Acute injections of the NMDA receptor antagonist memantine rescue performance deficits of the Ts65Dn mouse model of Down syndrome on a fear conditioning test. Neuropsychopharmacology 33, 1624–1632 (2008).

    Article  CAS  PubMed  Google Scholar 

  151. Lockrow, J., Boger, H., Bimonte-Nelson, H. & Granholm, A. C. Effects of long-term memantine on memory and neuropathology in Ts65Dn mice, a model for Down syndrome. Behav. Brain Res. 221, 610–622 (2011).

    Article  CAS  PubMed  Google Scholar 

  152. Rueda, N. et al. Memantine normalizes several phenotypic features in the Ts65Dn mouse model of Down syndrome. J. Alzheimers Dis. 21, 277–290 (2010).

    Article  CAS  PubMed  Google Scholar 

  153. Marvanova, M. et al. The neuroprotective agent memantine induces brain-derived neurotrophic factor and trkB receptor expression in rat brain. Mol. Cell. Neurosci. 18, 247–258 (2001).

    Article  CAS  PubMed  Google Scholar 

  154. Westmark, C. J. et al. Reversal of fragile X phenotypes by manipulation of AβPP/Aβ levels in Fmr1KO mice. PLoS ONE 6, e26549 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Lorenzi, H. A. & Reeves, R. H. Hippocampal hypocellularity in the Ts65Dn mouse originates early in development. Brain Res. 1104, 153–159 (2006).

    Article  CAS  PubMed  Google Scholar 

  156. Bianchi, P. et al. Early pharmacotherapy restores neurogenesis and cognitive performance in the Ts65Dn mouse model for Down syndrome. J. Neurosci. 30, 8769–8779 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Roper, R. J. et al. Defective cerebellar response to mitogenic Hedgehog signaling in Down [corrected] syndrome mice. Proc. Natl Acad. Sci. USA 103, 1452–1456 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Delcroix, J. D. et al. Trafficking the NGF signal: implications for normal and degenerating neurons. Prog. Brain Res. 146, 3–23 (2004).

    CAS  PubMed  Google Scholar 

  159. Salehi, A., Faizi, M., Belichenko, P. V. & Mobley, W. C. Using mouse models to explore genotype-phenotype relationship in Down syndrome. Ment. Retard. Dev. Disabil. Res. Rev. 13, 207–214 (2007).

    Article  PubMed  Google Scholar 

  160. Tejedor, F. J. & Hammerle, B. MNB/DYRK1A as a multiple regulator of neuronal development. FEBS J. 278, 223–235 (2011).

    Article  CAS  PubMed  Google Scholar 

  161. Hammerle, B. et al. Transient expression of Mnb/Dyrk1a couples cell cycle exit and differentiation of neuronal precursors by inducing p27KIP1 expression and suppressing NOTCH signaling. Development 138, 2543–2554 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Yabut, O., Domogauer, J. & D'Arcangelo, G. Dyrk1A overexpression inhibits proliferation and induces premature neuronal differentiation of neural progenitor cells. J. Neurosci. 30, 4004–4014 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Ortiz-Abalia, J. et al. Targeting Dyrk1A with AAVshRNA attenuates motor alterations in TgDyrk1A, a mouse model of Down syndrome. Am. J. Hum. Genet. 83, 479–488 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Guedj, F. et al. Green tea polyphenols rescue of brain defects induced by overexpression of DYRK1A. PLoS ONE 4, e4606 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Gockler, N. et al. Harmine specifically inhibits protein kinase DYRK1A and interferes with neurite formation. FEBS J. 276, 6324–6337 (2009).

    Article  CAS  PubMed  Google Scholar 

  166. Mazur-Kolecka, B. et al. Effect of DYRK1A activity inhibition on development of neuronal progenitors isolated from Ts65Dn mice. J. Neurosci. Res. 90, 999–1010 (2012).

    Article  CAS  PubMed  Google Scholar 

  167. Kamenetz, F. et al. APP processing and synaptic function. Neuron 37, 925–937 (2003).

    Article  CAS  PubMed  Google Scholar 

  168. Netzer, W. J. et al. Lowering β-amyloid levels rescues learning and memory in a Down syndrome mouse model. PLoS ONE 5, e10943 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Chakrabarti, L. et al. Olig1 and Olig2 triplication causes developmental brain defects in Down syndrome. Nature Neurosci. 13, 927–934 (2010). The authors identified OLIG1 and OLIG2 as important players in the developmental alterations, leading to increased inhibition in Down syndrome.

    Article  CAS  PubMed  Google Scholar 

  170. Haydar, T. F. & Reeves, R. H. Trisomy 21 and early brain development. Trends Neurosci. 35, 81–91 (2012).

    Article  CAS  PubMed  Google Scholar 

  171. Edgin, J. O. et al. Development and validation of the Arizona Cognitive Test Battery for Down syndrome. J. Neurodev Disord. 2, 149–164 (2010). The Arizona Cognitive Test Battery is the first serious attempt to create a neuropsychological exploration screen that is specific for Down syndrome.

    Article  PubMed  PubMed Central  Google Scholar 

  172. Nadel, L. Down's syndrome: a genetic disorder in biobehavioral perspective. Genes Brain Behav. 2, 156–166 (2003).

    Article  CAS  PubMed  Google Scholar 

  173. Ponting, C. P. & Belgard, T. G. Transcribed dark matter: meaning or myth? Hum. Mol. Genet. 19, R162–R168 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Elton, T. S., Sansom, S. E. & Martin, M. M. Trisomy-21 gene dosage over-expression of miRNAs results in the haploinsufficiency of specific target proteins. RNA Biol. 7, 540–547 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Senti, K. A. & Brennecke, J. The piRNA pathway: a fly's perspective on the guardian of the genome. Trends Genet. 26, 499–509 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Singh Sandhu, K., Li, G., Sung, W. K. & Ruan, Y. Chromatin interaction networks and higher order architectures of eukaryotic genomes. J. Cell. Biochem. 112, 2218–2221 (2011).

    Article  CAS  PubMed  Google Scholar 

  177. Reinholdt, L. G. et al. Meiotic behavior of aneuploid chromatin in mouse models of Down syndrome. Chromosoma 118, 723–736 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Cramer, N. & Galdzicki, Z. From abnormal hippocampal synaptic plasticity in down syndrome mouse models to cognitive disability in down syndrome. Neural Plast. 2012, 101542 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This Review is dedicated to all people with Down syndrome. I also dedicate it to M. Flórez and E. Bishop, and I offer a special thank you to J. Flórez, who is the professor that directed me to the field of Down syndrome research. I thank M. Martínez de Lagrán, G. Azkona and G. Arqué for their contributions to putting together the movies, and D. D'Amico for his contribution to figure 1. I apologize to all those colleagues whose work could not be cited directly in the manuscript due to space constraints. The work from my laboratory that is mentioned here was possible thanks to grants and contributions from the Jerôme Lejeune Foundation, Fundació Catalana Síndrome de Down, the Catalan Government (2009SGR1313), Spanish Ministry of Education and Sciences (SAF2007-60827, SAF2007-31093-E and SAF2010-16427), EU/FIS (PS09102673), CureFXS, ERARare, Fundación Ramón Areces, Alicia Koplowtiz, Marató TV3 and the Centre for Biomedical Network Research on Rare Diseases.

Author information

Authors and Affiliations

Authors

Ethics declarations

Competing interests

The author declares no competing financial interests.

Supplementary information

Supplementary information S1 (movie)

The Tapping test is part of the Wechsler Memory Scale-Revised and it is an adaptation of the Corsi-Block Tapping test (Orsini, A. Corsi’s block-tapping test: standardization and concurrent validity with WISC-R for children aged 11 to 16. Percept. Mot. Skills 79, 1547–1554 (1994)). It allows the study of the hippocampus-dependent visual-spatial declarative memory but also reflects attentional capacity, visual perceptive organization and executive functioning. This test is broadly used to define these specific abilities in different intellectual disability syndromes. Even though more sophisticated techniques are used now and computer versions of this test are available, the original version still provides very useful information. Briefly, the participants are presented in series with 'n' elements. In the absence of any difference between the shape and colour of the cubes, the spatial component is the most relevant. Sequences to be reproduced are randomly selected through a computer program, taking into account that the different spatial configurations provide a similar level of difficulty. Spatial memory is assessed by analysing the errors in reproducing the correct sequence of element taps. In general, the best recalled items in the series are the initial and final items (primacy and recency effect). Movie S1 shows a correct reproduction of the series by a healthy volunteer after the experimenter’s demonstration. The numbers are shown to the experimenter but are not visible for the participant. (MP4 314 kb)

Supplementary information S2 (movie)

Movie S2 shows the same series demonstrated by the experimenter in Movie S1, but here the healthy volunteer is not reproducing the series correctly. Instead the volunteer is demonstrating a typical visuospatial error, not maintaining the spatial configuration presented. Random errors or inclusion of an element that is not present in the series are also frequent in populations with intellectual disability. (MP4 364 kb)

Supplementary information S3 (movie)

Neuritogenesis in wild-type mice. The movie shows phase contrast time-lapse experiments performed to analyse axon growth in EGFP-transfected cultured cortical neurons derived from a 17.5-day-old wild-type mouse embryo (Martinez de Lagran, M. et al. Dyrk1A influences neuronal morphogenesis through regulation of cytoskeletal dynamics in mammalian cortical neurons. Cereb. Cortex 2 Jan 2012 (doi: 10.1093/cercor/bhr362)). Images were acquired at DIV 1 every 5 min for a period of 14 hours using 40 ms integration time to record stage coordinates of suitable axonal growth cones and analysed with particle track plugging of Image J software. (AVI 64555 kb)

Supplementary information S4 (movie)

Neuritogenesis in Down syndrome. The movie shows phase contrast time-lapse experiments performed to analyse axon growth in EGFP-transfected cultured cortical neurons derived from a 17.5-day-old Dyrk1A-overexpressing (TgDyrk1A) embryo. Images were acquired as described for Movie S3. Note that the axonal behaviour is different in transgenic neurons, which showed a significant reduction in the distance travelled by the axon leading to reduced axonal elongation, a phenotype that is also detected in Down syndrome (for detailed explanation, see Martinez de Lagran, M. et al. Dyrk1A influences neuronal morphogenesis through regulation of cytoskeletal dynamics in mammalian cortical neurons. Cereb. Cortex 2 Jan 2012 (doi: 10.1093/cercor/bhr362)). TgDyrk1A neurons presented with shorter terminal segments and less complex dendritic arbors with fewer dendrites, branch points and terminal segments. Mature synapse formation is reduced in the TgDyrk1A mouse, where filopodia-like spines are more abundant and mature spines are reduced in number (Martinez de Lagran, M. et al. Dyrk1A influences neuronal morphogenesis through regulation of cytoskeletal dynamics in mammalian cortical neurons. Cereb. Cortex 2 Jan 2012 (doi: 10.1093/cercor/bhr362); Popov, V. I., Kleschevnikov, A. M., Klimenko, O. A., Stewart, M. G. & Belichenko, P. V. Three-dimensional synaptic ultrastructure in the dentate gyrus and hippocampal area CA3 in the Ts65Dn mouse model of Down syndrome. J. Comp. Neurol. 519, 1338–1354 (2011); Tejedor, F. J. & Hammerle, B. MNB/DYRK1A as a multiple regulator of neuronal development. FEBS J. 278, 223–235 (2011); Belichenko, P. V. et al. Synaptic structural abnormalities in the Ts65Dn mouse model of Down Syndrome. J. Comp. Neurol. 480, 281–298 (2004)). (AVI 44186 kb)

Related links

Related links

FURTHER INFORMATION

Mara Dierssen's homepage

Down Syndrome International: Research and Practice

Down Syndrome Research and Treatment Foundation

Gene Function and Pathway Databases — GFuncPathdb

The HSA21 expression map initiative: a gene expression map of HSA21 orthologues in the mouse

Jerôme Lejeune Foundation

The National Down syndrome Congress

Online Mendelian Inheritance in Man: Down syndrome

Glossary

Intellectual disability

A disability that is characterized by significant limitations both in intellectual functioning and in adaptive behaviour.

Working memory

A system that is involved in the temporary storage and ongoing maintenance of information.

Long-term memory

A memory system for more permanently storing, managing and retrieving information for later use.

Explicit memory

This is the conscious processing of information to remember it following a delay.

Implicit memory

This comprises an unconscious, slower learning system, in which a previous experience influences current behaviour without consciousness of the first episode.

Brachycephaly

A condition in which an individual has an abnormally broad and short head, which occurs when the coronal sutures close prematurely.

Plasticity

This is defined as the capacity of the nervous system to modify its structural and functional organization as a result of experience.

Cognition

This is considered to be the process or processes whereby an organism gains knowledge or becomes aware of events or objects in its environment and uses that knowledge for comprehension and problem solving.

Small non-coding RNAs

These are regulatory genomic elements that are 18–30 nucleotides in length and include microRNAs, PIWI-interacting RNAs and endogenous small interfering RNAs.

Cerebral cortex

This is the outermost layer of the cerebral hemispheres of the brain and is largely responsible for all forms of conscious experience, including perception, emotion, thought and planning.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Dierssen, M. Down syndrome: the brain in trisomic mode. Nat Rev Neurosci 13, 844–858 (2012). https://doi.org/10.1038/nrn3314

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrn3314

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing