51
|
Bhutta MF, Cheeseman MT, Herault Y, Yu YE, Brown SDM. Surveying the Down syndrome mouse model resource identifies critical regions responsible for chronic otitis media. Mamm Genome 2013; 24:439-45. [PMID: 24068166 PMCID: PMC3843744 DOI: 10.1007/s00335-013-9475-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 08/15/2013] [Indexed: 02/06/2023]
Abstract
Chronic otitis media (OM) is common in Down syndrome (DS), but underlying aetiology is unclear. We analysed the entire available mouse resource of partial trisomy models of DS looking for histological evidence of chronic middle-ear inflammation. We found a highly penetrant OM in the Dp(16)1Yey mouse, which carries a complete trisomy of MMU16. No OM was found in the Dp(17)1Yey mouse or the Dp(10)1Yey mouse, suggesting disease loci are located only on MMU16. The Ts1Cje, Ts1RhR, Ts2Yah, and Ts65Dn trisomies and the transchomosomic Tc1 mouse did not develop OM. On the basis of these findings, we propose a two-locus model for chronic middle-ear inflammation in DS, based upon epistasis of the regions of HSA21 not in trisomy in the Tc1 mouse. We also conclude that environmental factors likely play an important role in disease onset.
Collapse
Affiliation(s)
- Mahmood F Bhutta
- Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Room 6607 Level 6, Headley Way, Oxford, OX3 9DU, UK,
| | | | | | | | | |
Collapse
|
52
|
Weaver DA, Nestor-Kalinoski AL, Craig K, Gorris M, Parikh T, Mabry H, Allison DC. Corrections for mRNA extraction and sample normalization errors find increased mRNA levels may compensate for cancer haplo-insufficiency. Genes Chromosomes Cancer 2013; 53:194-210. [PMID: 24327546 PMCID: PMC4237174 DOI: 10.1002/gcc.22133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 11/08/2013] [Accepted: 11/11/2013] [Indexed: 01/22/2023] Open
Abstract
The relative mRNA levels of differentially expressed (DE) and housekeeping (HK) genes of six aneuploid cancer lines with large-scale genomic changes identified by SNP/SKY analysis were compared with similar genes in diploid cells. The aneuploid cancer lines had heterogeneous genomic landscapes with subdiploid, diploid, and supradiploid regions and higher overall gene copy numbers compared with diploid cells. The mRNA levels of the haploid, diploid, and triploid HK genes were found to be higher after correction of easily identifiable mRNA measurement errors. Surprisingly, diploid and aneuploid HK gene mRNA levels were the same by standard expression array analyses, despite the higher copy numbers of the cancer cell HK genes. This paradoxical result proved to be due to inaccurate inputs of true intra-cellular mRNAs for analysis. These errors were corrected by analyzing the expression intensities of DE and HK genes in mRNAs extracted from equal cell numbers (50:50) of intact cancer cell and lymphocyte mixtures. Correction for both mRNA extraction/sample normalization errors and total gene copy numbers found the SUIT-2 and PC-3 cell lines' cancer genes both had ∼50% higher mRNA levels per single allele than lymphocyte gene alleles. These increased mRNA levels for single transcribed cancer alleles may restore functional mRNA levels to cancer genes rendered haplo-insufficient by the genetic instability of cancer. © 2013 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- David A Weaver
- Program in Bioinformatics and Proteomics/Genomics, The University of Toledo, College of Medicine and Life Sciences, Toledo, OH
| | | | | | | | | | | | | |
Collapse
|
53
|
Mouton-Liger F, Sahún I, Collin T, Lopes Pereira P, Masini D, Thomas S, Paly E, Luilier S, Même S, Jouhault Q, Bennaï S, Beloeil JC, Bizot JC, Hérault Y, Dierssen M, Créau N. Developmental molecular and functional cerebellar alterations induced by PCP4/PEP19 overexpression: implications for Down syndrome. Neurobiol Dis 2013; 63:92-106. [PMID: 24291518 DOI: 10.1016/j.nbd.2013.11.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 11/05/2013] [Accepted: 11/19/2013] [Indexed: 11/28/2022] Open
Abstract
PCP4/PEP19 is a modulator of Ca(2+)-CaM signaling. In the brain, it is expressed in a very specific pattern in postmitotic neurons. In particular, Pcp4 is highly expressed in the Purkinje cell, the sole output neuron of the cerebellum. PCP4, located on human chromosome 21, is present in three copies in individuals with Down syndrome (DS). In a previous study using a transgenic mouse model (TgPCP4) to evaluate the consequences of 3 copies of this gene, we found that PCP4 overexpression induces precocious neuronal differentiation during mouse embryogenesis. Here, we report combined analyses of the cerebellum at postnatal stages (P14 and adult) in which we identified age-related molecular, electrophysiological, and behavioral alterations in the TgPCP4 mouse. While Pcp4 overexpression at P14 induces an earlier neuronal maturation, at adult stage it induces increase in cerebellar CaMK2alpha and in cerebellar LTD, as well as learning impairments. We therefore propose that PCP4 contributes significantly to the development of Down syndrome phenotypes through molecular and functional changes.
Collapse
Affiliation(s)
- François Mouton-Liger
- Univ Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, EAC4413 CNRS, Paris, France
| | - Ignasi Sahún
- Cellular and Systems Biology, Systems Biology Programme, Center for Genomic Regulation (CRG); Universitat Pompeu Fabra (UPF); Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER): Dr. Aiguader, 88, 08003 Barcelona, Spain
| | - Thibault Collin
- CNRS UMR8118, Brain Physiology Laboratory, Universite Paris-Descartes, Centre universitaire des Saints-Pères, 45 Rue des Saints-Pères, 75270 Paris Cedex 06, France
| | - Patricia Lopes Pereira
- Transgenese et Archivage Animaux Modèles, TAAM, CNRS, UPS44, 3B rue de la Férollerie, 45071 Orléans, France
| | - Debora Masini
- Cellular and Systems Biology, Systems Biology Programme, Center for Genomic Regulation (CRG); Universitat Pompeu Fabra (UPF); Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER): Dr. Aiguader, 88, 08003 Barcelona, Spain
| | - Sophie Thomas
- Univ Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, EAC4413 CNRS, Paris, France
| | - Evelyne Paly
- Univ Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, EAC4413 CNRS, Paris, France
| | - Sabrina Luilier
- Key-Obs SAS, 13 avenue Buffon, 45071 Orléans Cedex 2, France
| | - Sandra Même
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Orléans, France
| | - Quentin Jouhault
- Univ Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, EAC4413 CNRS, Paris, France
| | - Soumia Bennaï
- Univ Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, EAC4413 CNRS, Paris, France
| | | | | | - Yann Hérault
- Transgenese et Archivage Animaux Modèles, TAAM, CNRS, UPS44, 3B rue de la Férollerie, 45071 Orléans, France; Institut Clinique de la Souris, ICS, 1 rue Laurent Fries, 67404 Illkirch, France; Institut de Génétique Biologie Moléculaire et Cellulaire, Translational medicine and Neuroscience program, IGBMC, CNRS, INSERM, Université de Strasbourg, UMR7104, UMR964, 1 rue Laurent Fries, 67404 Illkirch, France
| | - Mara Dierssen
- Cellular and Systems Biology, Systems Biology Programme, Center for Genomic Regulation (CRG); Universitat Pompeu Fabra (UPF); Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER): Dr. Aiguader, 88, 08003 Barcelona, Spain
| | - Nicole Créau
- Univ Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, EAC4413 CNRS, Paris, France.
| |
Collapse
|
54
|
Structural variation-associated expression changes are paralleled by chromatin architecture modifications. PLoS One 2013; 8:e79973. [PMID: 24265791 PMCID: PMC3827143 DOI: 10.1371/journal.pone.0079973] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 10/07/2013] [Indexed: 01/04/2023] Open
Abstract
Copy number variants (CNVs) influence the expression of genes that map not only within the rearrangement, but also to its flanks. To assess the possible mechanism(s) underlying this “neighboring effect”, we compared intrachromosomal interactions and histone modifications in cell lines of patients affected by genomic disorders and control individuals. Using chromosome conformation capture (4C-seq), we observed that a set of genes flanking the Williams-Beuren Syndrome critical region (WBSCR) were often looping together. The newly identified interacting genes include AUTS2, mutations of which are associated with autism and intellectual disabilities. Deletion of the WBSCR disrupts the expression of this group of flanking genes, as well as long-range interactions between them and the rearranged interval. We also pinpointed concomitant changes in histone modifications between samples. We conclude that large genomic rearrangements can lead to chromatin conformation changes that extend far away from the structural variant, thereby possibly modulating expression globally and modifying the phenotype. GEO Series accession number: GSE33784, GSE33867.
Collapse
|
55
|
Gene expression analysis of induced pluripotent stem cells from aneuploid chromosomal syndromes. BMC Genomics 2013; 14 Suppl 5:S8. [PMID: 24564826 PMCID: PMC3852284 DOI: 10.1186/1471-2164-14-s5-s8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background Human aneuploidy is the leading cause of early pregnancy loss, mental retardation, and multiple congenital anomalies. Due to the high mortality associated with aneuploidy, the pathophysiological mechanisms of aneuploidy syndrome remain largely unknown. Previous studies focused mostly on whether dosage compensation occurs, and the next generation transcriptomics sequencing technology RNA-seq is expected to eventually uncover the mechanisms of gene expression regulation and the related pathological phenotypes in human aneuploidy. Results Using next generation transcriptomics sequencing technology RNA-seq, we profiled the transcriptomes of four human aneuploid induced pluripotent stem cell (iPSC) lines generated from monosomy × (Turner syndrome), trisomy 8 (Warkany syndrome 2), trisomy 13 (Patau syndrome), and partial trisomy 11:22 (Emanuel syndrome) as well as two umbilical cord matrix iPSC lines as euploid controls to examine how phenotypic abnormalities develop with aberrant karyotype. A total of 466 M (50-bp) reads were obtained from the six iPSC lines, and over 13,000 mRNAs were identified by gene annotation. Global analysis of gene expression profiles and functional analysis of differentially expressed (DE) genes were implemented. Over 5000 DE genes are determined between aneuploidy and euploid iPSCs respectively while 9 KEGG pathways are overlapped enriched in four aneuploidy samples. Conclusions Our results demonstrate that the extra or missing chromosome has extensive effects on the whole transcriptome. Functional analysis of differentially expressed genes reveals that the genes most affected in aneuploid individuals are related to central nervous system development and tumorigenesis.
Collapse
|
56
|
Granese B, Scala I, Spatuzza C, Valentino A, Coletta M, Vacca RA, De Luca P, Andria G. Validation of microarray data in human lymphoblasts shows a role of the ubiquitin-proteasome system and NF-kB in the pathogenesis of Down syndrome. BMC Med Genomics 2013; 6:24. [PMID: 23830204 PMCID: PMC3717290 DOI: 10.1186/1755-8794-6-24] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 06/29/2013] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Down syndrome (DS) is a complex disorder caused by the trisomy of either the entire, or a critical region of chromosome 21 (21q22.1-22.3). Despite representing the most common cause of mental retardation, the molecular bases of the syndrome are still largely unknown. METHODS To better understand the pathogenesis of DS, we analyzed the genome-wide transcription profiles of lymphoblastoid cell lines (LCLs) from six DS and six euploid individuals and investigated differential gene expression and pathway deregulation associated with trisomy 21. Connectivity map and PASS-assisted exploration were used to identify compounds whose molecular signatures counteracted those of DS lymphoblasts and to predict their therapeutic potential. An experimental validation in DS LCLs and fetal fibroblasts was performed for the most deregulated GO categories, i.e. the ubiquitin mediated proteolysis and the NF-kB cascade. RESULTS We show, for the first time, that the level of protein ubiquitination is reduced in human DS cell lines and that proteasome activity is increased in both basal conditions and oxidative microenvironment. We also provide the first evidence that NF-kB transcription levels, a paradigm of gene expression control by ubiquitin-mediated degradation, is impaired in DS due to reduced IkB-alfa ubiquitination, increased NF-kB inhibitor (IkB-alfa) and reduced p65 nuclear fraction. Finally, the DSCR1/DYRK1A/NFAT genes were analysed. In human DS LCLs, we confirmed the presence of increased protein levels of DSCR1 and DYRK1A, and showed that the levels of the transcription factor NFATc2 were decreased in DS along with a reduction of its nuclear translocation upon induction of calcium fluxes. CONCLUSIONS The present work offers new perspectives to better understand the pathogenesis of DS and suggests a rationale for innovative approaches to treat some pathological conditions associated to DS.
Collapse
Affiliation(s)
- Barbara Granese
- Department of Pediatrics, Federico II University, Naples 80131, Italy
| | - Iris Scala
- Department of Pediatrics, Federico II University, Naples 80131, Italy
| | - Carmen Spatuzza
- Department of Biotechnological Sciences, Federico II University, Naples 80131, Italy
| | - Anna Valentino
- Department of Pediatrics, Federico II University, Naples 80131, Italy
| | - Marcella Coletta
- Department of Pediatrics, Federico II University, Naples 80131, Italy
| | - Rosa Anna Vacca
- Institute of Biomembranes and Bioenergetics, National Council of Research, Bari 70126, Italy
| | - Pasquale De Luca
- Stazione Zoologica “A. Dohrn”, c/o BioGeM, Via Camporeale, Ariano Irpino 83031, Italy
| | - Generoso Andria
- Department of Pediatrics, Federico II University, Naples 80131, Italy
| |
Collapse
|
57
|
Sailani MR, Makrythanasis P, Valsesia A, Santoni FA, Deutsch S, Popadin K, Borel C, Migliavacca E, Sharp AJ, Duriaux Sail G, Falconnet E, Rabionet K, Serra-Juhé C, Vicari S, Laux D, Grattau Y, Dembour G, Megarbane A, Touraine R, Stora S, Kitsiou S, Fryssira H, Chatzisevastou-Loukidou C, Kanavakis E, Merla G, Bonnet D, Pérez-Jurado LA, Estivill X, Delabar JM, Antonarakis SE. The complex SNP and CNV genetic architecture of the increased risk of congenital heart defects in Down syndrome. Genome Res 2013; 23:1410-21. [PMID: 23783273 PMCID: PMC3759718 DOI: 10.1101/gr.147991.112] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Congenital heart defect (CHD) occurs in 40% of Down syndrome (DS) cases. While carrying three copies of chromosome 21 increases the risk for CHD, trisomy 21 itself is not sufficient to cause CHD. Thus, additional genetic variation and/or environmental factors could contribute to the CHD risk. Here we report genomic variations that in concert with trisomy 21, determine the risk for CHD in DS. This case-control GWAS includes 187 DS with CHD (AVSD = 69, ASD = 53, VSD = 65) as cases, and 151 DS without CHD as controls. Chromosome 21–specific association studies revealed rs2832616 and rs1943950 as CHD risk alleles (adjusted genotypic P-values <0.05). These signals were confirmed in a replication cohort of 92 DS-CHD cases and 80 DS-without CHD (nominal P-value 0.0022). Furthermore, CNV analyses using a customized chromosome 21 aCGH of 135K probes in 55 DS-AVSD and 53 DS-without CHD revealed three CNV regions associated with AVSD risk (FDR ≤ 0.05). Two of these regions that are located within the previously identified CHD region on chromosome 21 were further confirmed in a replication study of 49 DS-AVSD and 45 DS- without CHD (FDR ≤ 0.05). One of these CNVs maps near the RIPK4 gene, and the second includes the ZBTB21 (previously ZNF295) gene, highlighting the potential role of these genes in the pathogenesis of CHD in DS. We propose that the genetic architecture of the CHD risk of DS is complex and includes trisomy 21, and SNP and CNV variations in chromosome 21. In addition, a yet-unidentified genetic variation in the rest of the genome may contribute to this complex genetic architecture.
Collapse
Affiliation(s)
- M Reza Sailani
- Department of Genetic Medicine and Development, University of Geneva, Geneva 1211, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Jin S, Lee YK, Lim YC, Zheng Z, Lin XM, Ng DPY, Holbrook JD, Law HY, Kwek KYC, Yeo GSH, Ding C. Global DNA hypermethylation in down syndrome placenta. PLoS Genet 2013; 9:e1003515. [PMID: 23754950 PMCID: PMC3675012 DOI: 10.1371/journal.pgen.1003515] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2012] [Accepted: 04/04/2013] [Indexed: 11/19/2022] Open
Abstract
Down syndrome (DS), commonly caused by an extra copy of chromosome 21 (chr21), occurs in approximately one out of 700 live births. Precisely how an extra chr21 causes over 80 clinically defined phenotypes is not yet clear. Reduced representation bisulfite sequencing (RRBS) analysis at single base resolution revealed DNA hypermethylation in all autosomes in DS samples. We hypothesize that such global hypermethylation may be mediated by down-regulation of TET family genes involved in DNA demethylation, and down-regulation of REST/NRSF involved in transcriptional and epigenetic regulation. Genes located on chr21 were up-regulated by an average of 53% in DS compared to normal villi, while genes with promoter hypermethylation were modestly down-regulated. DNA methylation perturbation was conserved in DS placenta villi and in adult DS peripheral blood leukocytes, and enriched for genes known to be causally associated with DS phenotypes. Our data suggest that global epigenetic changes may occur early in development and contribute to DS phenotypes.
Collapse
Affiliation(s)
- Shengnan Jin
- Growth, Development and Metabolism Program, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore
| | - Yew Kok Lee
- Growth, Development and Metabolism Program, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore
| | - Yen Ching Lim
- Growth, Development and Metabolism Program, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore
| | - Zejun Zheng
- Growth, Development and Metabolism Program, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore
| | - Xueqin Michelle Lin
- Growth, Development and Metabolism Program, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore
| | - Desmond P. Y. Ng
- Growth, Development and Metabolism Program, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore
| | - Joanna D. Holbrook
- Growth, Development and Metabolism Program, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore
| | | | | | | | - Chunming Ding
- Growth, Development and Metabolism Program, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore
- * E-mail:
| |
Collapse
|
59
|
Cho CKJ, Drabovich AP, Karagiannis GS, Martínez-Morillo E, Dason S, Dimitromanolakis A, Diamandis EP. Quantitative proteomic analysis of amniocytes reveals potentially dysregulated molecular networks in Down syndrome. Clin Proteomics 2013; 10:2. [PMID: 23394617 PMCID: PMC3626793 DOI: 10.1186/1559-0275-10-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 01/18/2013] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Down syndrome (DS), caused by an extra copy of chromosome 21, affects 1 in 750 live births and is characterized by cognitive impairment and a constellation of congenital defects. Currently, little is known about the molecular pathogenesis and no direct genotype-phenotype relationship has yet been confirmed. Since DS amniocytes are expected to have a distinct biological behaviour compared to normal amniocytes, we hypothesize that relative quantification of proteins produced from trisomy and euploid (chromosomally normal) amniocytes will reveal dysregulated molecular pathways. RESULTS Chromosomally normal- and Trisomy 21-amniocytes were quantitatively analyzed by using Stable Isotope Labeling of Amino acids in Cell culture and tandem mass spectrometry. A total of 4919 unique proteins were identified from the supernatant and cell lysate proteome. More specifically, 4548 unique proteins were identified from the lysate, and 91% of these proteins were quantified based on MS/MS spectra ratios of peptides containing isotope-labeled amino acids. A total of 904 proteins showed significant differential expression and were involved in 25 molecular pathways, each containing a minimum of 16 proteins. Sixty of these proteins consistently showed aberrant expression from trisomy 21 affected amniocytes, indicating their potential role in DS pathogenesis. Nine proteins were analyzed with a multiplex selected reaction monitoring assay in an independent set of Trisomy 21-amniocyte samples and two of them (SOD1 and NES) showed a consistent differential expression. CONCLUSIONS The most extensive proteome of amniocytes and amniotic fluid has been generated and differentially expressed proteins from amniocytes with Trisomy 21 revealed molecular pathways that seem to be most significantly affected by the presence of an extra copy of chromosome 21.
Collapse
Affiliation(s)
- Chan-Kyung J Cho
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| | | | | | | | | | | | | |
Collapse
|
60
|
Tang YC, Amon A. Gene copy-number alterations: a cost-benefit analysis. Cell 2013; 152:394-405. [PMID: 23374337 PMCID: PMC3641674 DOI: 10.1016/j.cell.2012.11.043] [Citation(s) in RCA: 265] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 10/22/2012] [Accepted: 11/01/2012] [Indexed: 11/25/2022]
Abstract
Changes in DNA copy number, whether confined to specific genes or affecting whole chromosomes, have been identified as causes of diseases and developmental abnormalities and as sources of adaptive potential. Here, we discuss the costs and benefits of DNA copy-number alterations. Changes in DNA copy number are largely detrimental. Amplifications or deletions of specific genes can elicit discrete defects. Large-scale changes in DNA copy number can also cause detrimental phenotypes that are due to the cumulative effects of copy-number alterations of many genes simultaneously. On the other hand, studies in microorganisms show that DNA copy-number alterations can be beneficial, increasing survival under selective pressure. As DNA copy-number alterations underlie many human diseases, we will end with a discussion of gene copy-number changes as therapeutic targets.
Collapse
Affiliation(s)
- Yun-Chi Tang
- David H. Koch Institute for Integrative Cancer Research, Howard Hughes Medical Institute, Massachusetts Institute of Technology, 76-561, 500 Main Street, Cambridge, MA 02139, USA
| | - Angelika Amon
- David H. Koch Institute for Integrative Cancer Research, Howard Hughes Medical Institute, Massachusetts Institute of Technology, 76-561, 500 Main Street, Cambridge, MA 02139, USA
| |
Collapse
|
61
|
Spellman C, Ahmed MM, Dubach D, Gardiner KJ. Expression of trisomic proteins in Down syndrome model systems. Gene 2012; 512:219-25. [PMID: 23103828 DOI: 10.1016/j.gene.2012.10.051] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2012] [Revised: 09/06/2012] [Accepted: 10/09/2012] [Indexed: 12/21/2022]
Abstract
Down syndrome (DS) is the most common genetic aberration leading to intellectual disability. DS results from an extra copy of the long arm of human chromosome 21 (HSA21) and the increased expression of trisomic genes due to gene dosage. While expression in DS and DS models has been studied extensively at the RNA level, much less is known about expression of trisomic genes at the protein level. We have used quantitative Western blotting with antibodies to 20 proteins encoded by HSA21 to assess trisomic protein expression in lymphoblastoid cell lines (LCLs) from patients with DS and in brains from two mouse models of DS. These antibodies have recently become available and the 20 proteins largely have not been investigated previously for their potential contributions to the phenotypic features of DS. Twelve proteins had detectable expression in LCLs and three, CCT8, MX1 and PWP2, showed elevated levels in LCLs derived from patients with DS compared with controls. Antibodies against 15 proteins detected bands of appropriate sizes in lysates from mouse brain cortex. Genes for 12 of these proteins are trisomic in the Tc1 mouse model of DS, but only SIM2 and ZNF295 showed elevated expression in Tc1 cortex when compared with controls. Genes for eight of the 15 proteins are trisomic in the Ts65Dn mouse model of DS, but only ZNF294 was over expressed in cortex. Comparison of trisomic gene expression at the protein level with previous reports at the mRNA level showed many inconsistencies. These may be caused by natural inter-individual variability, differences in the age of mice analyzed, or post-transcriptional regulation of gene dosage effects. These antibodies provide resources for further investigation of the molecular basis of intellectual disability in DS.
Collapse
Affiliation(s)
- Claire Spellman
- Department of Pediatrics, Linda Crnic Institute for Down Syndrome, Intellectual and Developmental Disabilities Research Center, University of Colorado Denver School of Medicine, 12700 E 19th Avenue, Mail Stop 8608, Aurora, CO 80045, USA.
| | | | | | | |
Collapse
|
62
|
Trisomy 21-associated defects in human primitive hematopoiesis revealed through induced pluripotent stem cells. Proc Natl Acad Sci U S A 2012; 109:17573-8. [PMID: 23045704 DOI: 10.1073/pnas.1211175109] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Patients with Down syndrome (trisomy 21, T21) have hematologic abnormalities throughout life. Newborns frequently exhibit abnormal blood counts and a clonal preleukemia. Human T21 fetal livers contain expanded erythro-megakaryocytic precursors with enhanced proliferative capacity. The impact of T21 on the earliest stages of embryonic hematopoiesis is unknown and nearly impossible to examine in human subjects. We modeled T21 yolk sac hematopoiesis using human induced pluripotent stem cells (iPSCs). Blood progenitor populations generated from T21 iPSCs were present at normal frequency and proliferated normally. However, their developmental potential was altered with enhanced erythropoiesis and reduced myelopoiesis, but normal megakaryocyte production. These abnormalities overlap with those of T21 fetal livers, but also reflect important differences. Our studies show that T21 confers distinct developmental stage- and species-specific hematopoietic defects. More generally, we illustrate how iPSCs can provide insight into early stages of normal and pathological human development.
Collapse
|
63
|
Molecular and cellular alterations in Down syndrome: toward the identification of targets for therapeutics. Neural Plast 2012; 2012:171639. [PMID: 22848846 PMCID: PMC3403492 DOI: 10.1155/2012/171639] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 04/18/2012] [Accepted: 04/19/2012] [Indexed: 12/25/2022] Open
Abstract
Down syndrome is a complex disease that has challenged molecular and cellular research for more than 50 years. Understanding the molecular bases of morphological, cellular, and functional alterations resulting from the presence of an additional complete chromosome 21 would aid in targeting specific genes and pathways for rescuing some phenotypes. Recently, progress has been made by characterization of brain alterations in mouse models of Down syndrome. This review will highlight the main molecular and cellular findings recently described for these models, particularly with respect to their relationship to Down syndrome phenotypes.
Collapse
|
64
|
Abstract
Deviation from a balanced genome by either gain or loss of entire chromosomes is generally tolerated poorly in all eukaryotic systems studied to date. Errors in mitotic or meiotic cell division lead to aneuploidy, which places a burden of additional or insufficient gene products from the missegregated chromosomes on the daughter cells. The burden of aneuploidy often manifests itself as impaired fitness of individual cells and whole organisms, in which abnormal development is also characteristic. However, most human cancers, noted for their rapid growth, also display various levels of aneuploidy. Here we discuss the detrimental, potentially beneficial, and sometimes puzzling effects of aneuploidy on cellular and organismal fitness and tissue function as well as its role in diseases such as cancer and neurodegeneration.
Collapse
Affiliation(s)
- Jake J Siegel
- David H. Koch Institute for Integrative Cancer Research and Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA.
| | | |
Collapse
|
65
|
Raveau M, Lignon JM, Nalesso V, Duchon A, Groner Y, Sharp AJ, Dembele D, Brault V, Hérault Y. The App-Runx1 region is critical for birth defects and electrocardiographic dysfunctions observed in a Down syndrome mouse model. PLoS Genet 2012; 8:e1002724. [PMID: 22693452 PMCID: PMC3364940 DOI: 10.1371/journal.pgen.1002724] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 04/05/2012] [Indexed: 01/24/2023] Open
Abstract
Down syndrome (DS) leads to complex phenotypes and is the main genetic cause of birth defects and heart diseases. The Ts65Dn DS mouse model is trisomic for the distal part of mouse chromosome 16 and displays similar features with post-natal lethality and cardiovascular defects. In order to better understand these defects, we defined electrocardiogram (ECG) with a precordial set-up, and we found conduction defects and modifications in wave shape, amplitudes, and durations in Ts65Dn mice. By using a genetic approach consisting of crossing Ts65Dn mice with Ms5Yah mice monosomic for the App-Runx1 genetic interval, we showed that the Ts65Dn viability and ECG were improved by this reduction of gene copy number. Whole-genome expression studies confirmed gene dosage effect in Ts65Dn, Ms5Yah, and Ts65Dn/Ms5Yah hearts and showed an overall perturbation of pathways connected to post-natal lethality (Coq7, Dyrk1a, F5, Gabpa, Hmgn1, Pde10a, Morc3, Slc5a3, and Vwf) and heart function (Tfb1m, Adam19, Slc8a1/Ncx1, and Rcan1). In addition cardiac connexins (Cx40, Cx43) and sodium channel sub-units (Scn5a, Scn1b, Scn10a) were found down-regulated in Ts65Dn atria with additional down-regulation of Cx40 in Ts65Dn ventricles and were likely contributing to conduction defects. All these data pinpoint new cardiac phenotypes in the Ts65Dn, mimicking aspects of human DS features and pathways altered in the mouse model. In addition they highlight the role of the App-Runx1 interval, including Sod1 and Tiam1, in the induction of post-natal lethality and of the cardiac conduction defects in Ts65Dn. These results might lead to new therapeutic strategies to improve the care of DS people.
Collapse
Affiliation(s)
- Matthieu Raveau
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, CNRS, INSERM, Université de Strasbourg, UMR7104, UMR964, Illkirch, France
| | - Jacques M. Lignon
- Immunologie et Embryologie Moléculaire, CNRS Université d'Orléans, UMR6218, Orléans, France
| | - Valérie Nalesso
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, CNRS, INSERM, Université de Strasbourg, UMR7104, UMR964, Illkirch, France
| | - Arnaud Duchon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, CNRS, INSERM, Université de Strasbourg, UMR7104, UMR964, Illkirch, France
| | - Yoram Groner
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Andrew J. Sharp
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Doulaye Dembele
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, CNRS, INSERM, Université de Strasbourg, UMR7104, UMR964, Illkirch, France
| | - Véronique Brault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, CNRS, INSERM, Université de Strasbourg, UMR7104, UMR964, Illkirch, France
| | - Yann Hérault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, CNRS, INSERM, Université de Strasbourg, UMR7104, UMR964, Illkirch, France
- Transgénèse et Archivage d'Animaux Modèles, CNRS, UPS44, Orléans, France
- Institut Clinique de la Souris, Illkirch, France
- * E-mail:
| |
Collapse
|
66
|
Hansen JS, Nygaard UC, Lyle R, Lovik M. Early Life Interventions to Prevent Allergy in the Offspring: The Role of Maternal Immunization and Postnatal Mucosal Allergen Exposure. Int Arch Allergy Immunol 2012; 158:261-75. [DOI: 10.1159/000332963] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Accepted: 09/07/2011] [Indexed: 02/05/2023] Open
|
67
|
The use of mouse models for understanding the biology of down syndrome and aging. Curr Gerontol Geriatr Res 2012; 2012:717315. [PMID: 22461792 PMCID: PMC3296169 DOI: 10.1155/2012/717315] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2011] [Accepted: 12/06/2011] [Indexed: 12/16/2022] Open
Abstract
Down syndrome is a complex condition caused by trisomy of human chromosome 21. The biology of aging may be different in individuals with Down syndrome; this is not well understood in any organism. Because of its complexity, many aspects of Down syndrome must be studied either in humans or in animal models. Studies in humans are essential but are limited for ethical and practical reasons. Fortunately, genetically altered mice can serve as extremely useful models of Down syndrome, and progress in their production and analysis has been remarkable. Here, we describe various mouse models that have been used to study Down syndrome. We focus on segmental trisomies of mouse chromosome regions syntenic to human chromosome 21, mice in which individual genes have been introduced, or mice in which genes have been silenced by targeted mutagenesis. We selected a limited number of genes for which considerable evidence links them to aspects of Down syndrome, and about which much is known regarding their function. We focused on genes important for brain and cognitive function, and for the altered cancer spectrum seen in individuals with Down syndrome. We conclude with observations on the usefulness of mouse models and speculation on future directions.
Collapse
|
68
|
Genomic determinants in the phenotypic variability of Down syndrome. PROGRESS IN BRAIN RESEARCH 2012; 197:15-28. [PMID: 22541286 DOI: 10.1016/b978-0-444-54299-1.00002-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Down syndrome caused by trisomy 21 is a collection of phenotypes with variable expressivity and penetrance. The significant advances in exploring the human genome now provide the tools to better understand the contribution of trisomy 21 in the different manifestations of Down syndrome, and the functional links between the genome variability and the phenotypic variability.
Collapse
|
69
|
Krishnamoorthy M, Gerwe BA, Scharer CD, Heimburg-Molinaro J, Gregory F, Nash RJ, Arumugham J, Usta SN, Eilertson CD, Stice SL, Nash RJ. GABRB3 gene expression increases upon ethanol exposure in human embryonic stem cells. J Recept Signal Transduct Res 2011; 31:206-13. [PMID: 21619448 DOI: 10.3109/10799893.2011.569723] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Ionotropic receptors are the target for most mood-defining compounds. Chronic exposure to ethanol (EtOH) alters receptor-mediated responses and the numbers of these channels and specific subunits; as well as induces anxiolytic, sedative, and anesthetic activity in the human brain. However, very little is known regarding the effects of EtOH on ionotropic receptor transcription during early human development (preimplantation). Using two separate human embryonic stem cell lines the study shows that low amounts of EtOH (20 mM) alters transcription of the ionotropic subunit GABRB3. Changes in ionotrophic receptor expression influence the central nervous system development and have been shown to produce brain abnormalities in animal models. These results suggest that low concentrations of EtOH can alter ionotropic receptor transcription during early human development (preimplantation), which may be a contributing factor to the neurological phenotypes seen in fetal alcohol spectrum disorder (FASD).
Collapse
|
70
|
von Kanel T, Gerber D, Wittwer CT, Hermann M, Gallati S. Detecting and resolving position-dependent temperature effects in real-time quantitative polymerase chain reaction. Anal Biochem 2011; 419:161-7. [DOI: 10.1016/j.ab.2011.08.041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Revised: 08/10/2011] [Accepted: 08/10/2011] [Indexed: 10/17/2022]
|
71
|
Brault V, Martin B, Costet N, Bizot JC, Hérault Y. Characterization of PTZ-induced seizure susceptibility in a down syndrome mouse model that overexpresses CSTB. PLoS One 2011; 6:e27845. [PMID: 22140471 PMCID: PMC3227573 DOI: 10.1371/journal.pone.0027845] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 10/26/2011] [Indexed: 12/27/2022] Open
Abstract
Down syndrome (DS) is a complex genetic syndrome characterized by intellectual disability, dysmorphism and variable additional physiological traits. Current research progress has begun to decipher the neural mechanisms underlying cognitive impairment, leading to new therapeutic perspectives. Pentylenetetrazol (PTZ) has recently been found to have positive effects on learning and memory capacities of a DS mouse model and is foreseen to treat DS patients. But PTZ is also known to be a convulsant drug at higher dose and DS persons are more prone to epileptic seizures than the general population. This raises concerns over what long-term effects of treatment might be in the DS population. The cause of increased propensity for epilepsy in the DS population and which Hsa21 gene(s) are implicated remain unknown. Among Hsa21 candidate genes in epilepsy, CSTB, coding for the cystein protease inhibitor cystatin B, is involved in progressive myoclonus epilepsy and ataxia in both mice and human. Thus we aim to evaluate the effect of an increase in Cstb gene dosage on spontaneous epileptic activity and susceptibility to PTZ-induced seizure. To this end we generated a new mouse model trisomic for Cstb by homologous recombination. We verified that increasing copy number of Cstb from Trisomy (Ts) to Tetrasomy (Tt) was driving overexpression of the gene in the brain, we checked transgenic animals for presence of locomotor activity and electroencephalogram (EEG) abnormalities characteristic of myoclonic epilepsy and we tested if those animals were prone to PTZ-induced seizure. Overall, the results of the analysis shows that an increase in Cstb does not induce any spontaneous epileptic activity and neither increase or decrease the propensity of Ts and Tt mice to myoclonic seizures suggesting that Ctsb dosage should not interfere with PTZ-treatment.
Collapse
Affiliation(s)
- Véronique Brault
- Department of Translational Medicine and Neurogenetics, Institut de Génétique Biologie Moléculaire et Cellulaire (IGBMC), Inserm U596, CNRS UMR7104, Université de Strasbourg, Illkirch, France
| | - Benoît Martin
- Inserm U642, Rennes, France
- Laboratoire Traitement du Signal et de l'Image, Université de Rennes 1, Rennes, France
| | - Nathalie Costet
- Inserm U642, Rennes, France
- Laboratoire Traitement du Signal et de l'Image, Université de Rennes 1, Rennes, France
| | | | - Yann Hérault
- Department of Translational Medicine and Neurogenetics, Institut de Génétique Biologie Moléculaire et Cellulaire (IGBMC), Inserm U596, CNRS UMR7104, Université de Strasbourg, Illkirch, France
- Transgenese et Archivage Animaux Modèles, TAAM, CNRS, UPS44, Orléans, France
- Institut Français Clinique de la Souris, GIE CERBM, Illkirch, France
| |
Collapse
|
72
|
Mouton-Liger F, Thomas S, Rattenbach R, Magnol L, Larigaldie V, Ledru A, Herault Y, Verney C, Créau N. PCP4 (PEP19) overexpression induces premature neuronal differentiation associated with Ca(2+) /calmodulin-dependent kinase II-δ activation in mouse models of Down syndrome. J Comp Neurol 2011; 519:2779-802. [PMID: 21491429 DOI: 10.1002/cne.22651] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Pcp4/pep19 is a modulator of Ca(2+) -CaM, a key molecule for calcium signaling, expressed in postmitotic neuroectoderm cells during mouse embryogenesis. The PCP4 gene is located on human chromosome 21 and is present in three copies in Down syndrome (DS). To evaluate the consequences of three copies of this gene on the development of these cells in the nervous system, we constructed a transgenic (TgPCP4) mouse model, with one copy of human PCP4, and investigated the effects in this model and in the Ts1Cje, a mouse model of DS. During embryogenesis, we analyzed 1) the level of pcp4 transcript and protein in the two models; 2) the extent of colabeling for markers of neuronal differentiation (βIII-tubulin, Map2c, calbindin, and calretinin) and pcp4 by immunofluorescence analysis and overall protein levels of these markers by Western blotting; and 3) the rate of activation of CaMKII, a Ca(2+) -CaM target, to evaluate the impact of pcp4 overexpression on the Ca(2+) -CaM signaling pathway. We showed that three copies of the pcp4 gene induced the overexpression of transcripts and proteins during embryogenesis. Pcp4 overexpression 1) induced precocious neuronal differentiation, as shown by the distribution and levels of early neuronal markers; and 2) was associated with an increase in CaMKIIδ activation, confirming involvement in neuronal differentiation in vivo via a Pcp4-Ca(2+) -CaM pathway. TgPCP4 and Ts1Cje mice developed similar modifications, demonstrating that these mechanisms may account for abnormal neuronal development in DS.
Collapse
Affiliation(s)
- François Mouton-Liger
- Functional Adaptive Biology (BFA), Centre National de la Recherche Scientifique (CNRS) EAC4413, Université Paris Diderot-Paris7, 75205 Paris Cedex 13, France
| | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Liu C, Morishima M, Yu T, Matsui SI, Zhang L, Fu D, Pao A, Costa AC, Gardiner KJ, Cowell JK, Nowak NJ, Parmacek MS, Liang P, Baldini A, Yu YE. Genetic analysis of Down syndrome-associated heart defects in mice. Hum Genet 2011; 130:623-32. [PMID: 21442329 PMCID: PMC3257027 DOI: 10.1007/s00439-011-0980-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Accepted: 03/07/2011] [Indexed: 01/22/2023]
Abstract
Human trisomy 21, the chromosomal basis of Down syndrome (DS), is the most common genetic cause of heart defects. Regions on human chromosome 21 (Hsa21) are syntenically conserved with three regions located on mouse chromosome 10 (Mmu10), Mmu16 and Mmu17. In this study, we have analyzed the impact of duplications of each syntenic region on cardiovascular development in mice and have found that only the duplication on Mmu16, i.e., Dp(16)1Yey, is associated with heart defects. Furthermore, we generated two novel mouse models carrying a 5.43-Mb duplication and a reciprocal deletion between Tiam1 and Kcnj6 using chromosome engineering, Dp(16Tiam1-Kcnj6)Yey/+ and Df(16Tiam1-Kcnj6)Yey/+, respectively, within the 22.9-Mb syntenic region on Mmu16. We found that Dp(16Tiam1-Kcnj6)Yey/+, but not Dp(16)1Yey/Df(16Tiam1-Kcnj6)Yey, resulted in heart defects, indicating that triplication of the Tiam1-Knj6 region is necessary and sufficient to cause DS-associated heart defects. Our transcriptional analysis of Dp(16Tiam1-Kcnj6)Yey/+ embryos confirmed elevated expression levels for the genes located in the Tiam-Kcnj6 region. Therefore, we established the smallest critical genomic region for DS-associated heart defects to lay the foundation for identifying the causative gene(s) for this phenotype.
Collapse
Affiliation(s)
- Chunhong Liu
- Children’s Guild Foundation Down Syndrome Research Program and Department of Cancer Genetics, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Masae Morishima
- Department of Anatomy and Developmental Biology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Tao Yu
- Children’s Guild Foundation Down Syndrome Research Program and Department of Cancer Genetics, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Sei-Ichi Matsui
- Children’s Guild Foundation Down Syndrome Research Program and Department of Cancer Genetics, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Li Zhang
- Children’s Guild Foundation Down Syndrome Research Program and Department of Cancer Genetics, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Dawei Fu
- Children’s Guild Foundation Down Syndrome Research Program and Department of Cancer Genetics, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Annie Pao
- Children’s Guild Foundation Down Syndrome Research Program and Department of Cancer Genetics, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Alberto C. Costa
- Division of Clinical Pharmacology and Toxicology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Katheleen J. Gardiner
- Department of Pediatrics, Intellectual and Developmental Disability Research Center, Human Medical Genetics and Neuroscience Programs, University of Colorado Denver, Aurora, CO 80045, USA
| | - John K. Cowell
- Children’s Guild Foundation Down Syndrome Research Program and Department of Cancer Genetics, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA
- MCG Cancer Center, School of Medicine, Medical College of Georgia, Augusta, GA 30912, USA
| | - Norma J. Nowak
- Children’s Guild Foundation Down Syndrome Research Program and Department of Cancer Genetics, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA
- New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY 14263, USA; Department of Cellular and Molecular Biology, Roswell Park Division of Graduate School, State University of New York at Buffalo, Buffalo, NY 14263, USA
| | - Michael S. Parmacek
- Departments of Cell and Developmental Biology and Medicine, University of Pennsylvania Cardiovascular Institute, Philadelphia, PA 19104, USA
| | - Ping Liang
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Antonio Baldini
- Institute of Biosciences and Technologies, Texas A&M University, Houston, TX 77843, USA; Institute of Genetics and Biophysics, National Research Council, 80131 Naples, Italy
| | - Y. Eugene Yu
- Children’s Guild Foundation Down Syndrome Research Program and Department of Cancer Genetics, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA
- New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY 14263, USA; Department of Cellular and Molecular Biology, Roswell Park Division of Graduate School, State University of New York at Buffalo, Buffalo, NY 14263, USA
| |
Collapse
|
74
|
Keck-Wherley J, Grover D, Bhattacharyya S, Xu X, Holman D, Lombardini ED, Verma R, Biswas R, Galdzicki Z. Abnormal microRNA expression in Ts65Dn hippocampus and whole blood: contributions to Down syndrome phenotypes. Dev Neurosci 2011; 33:451-67. [PMID: 22042248 PMCID: PMC3254042 DOI: 10.1159/000330884] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Accepted: 07/06/2011] [Indexed: 12/22/2022] Open
Abstract
Down syndrome (DS; trisomy 21) is one of the most common genetic causes of intellectual disability, which is attributed to triplication of genes located on chromosome 21. Elevated levels of several microRNAs (miRNAs) located on chromosome 21 have been reported in human DS heart and brain tissues. The Ts65Dn mouse model is the most investigated DS model with a triplicated segment of mouse chromosome 16 harboring genes orthologous to those on human chromosome 21. Using ABI TaqMan miRNA arrays, we found a set of miRNAs that were significantly up- or downregulated in the Ts65Dn hippocampus compared to euploid controls. Furthermore, miR-155 and miR-802 showed significant overexpression in the Ts65Dn hippocampus, thereby confirming results of previous studies. Interestingly, miR-155 and miR-802 were also overexpressed in the Ts65Dn whole blood but not in lung tissue. We also found overexpression of the miR-155 precursors, pri- and pre-miR-155 derived from the miR-155 host gene, known as B cell integration cluster, suggesting enhanced biogenesis of miR-155. Bioinformatic analysis revealed that neurodevelopment, differentiation of neuroglia, apoptosis, cell cycle, and signaling pathways including ERK/MAPK, protein kinase C, phosphatidylinositol 3-kinase, m-TOR and calcium signaling are likely targets of these miRNAs. We selected some of these potential gene targets and found downregulation of mRNA encoding Ship1, Mecp2 and Ezh2 in Ts65Dn hippocampus. Interestingly, the miR-155 target gene Ship1 (inositol phosphatase) was also downregulated in Ts65Dn whole blood but not in lung tissue. Our findings provide insights into miRNA-mediated gene regulation in Ts65Dn mice and their potential contribution to impaired hippocampal synaptic plasticity and neurogenesis, as well as hemopoietic abnormalities observed in DS.
Collapse
Affiliation(s)
- Jennifer Keck-Wherley
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, Md., USA
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Md., USA
| | - Deepak Grover
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Md., USA
| | - Sharmistha Bhattacharyya
- Department of Graduate School of Nursing, Uniformed Services University of the Health Sciences, Bethesda, Md., USA
| | - Xiufen Xu
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Md., USA
| | - Derek Holman
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Md., USA
| | - Eric D. Lombardini
- Department of Comparative Pathology Division, Veterinary Sciences Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Md., USA
| | - Ranjana Verma
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Md., USA
| | - Roopa Biswas
- Department of Graduate School of Nursing, Uniformed Services University of the Health Sciences, Bethesda, Md., USA
| | - Zygmunt Galdzicki
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Md., USA
| |
Collapse
|
75
|
Liu C, Belichenko PV, Zhang L, Fu D, Kleschevnikov AM, Baldini A, Antonarakis SE, Mobley WC, Yu YE. Mouse models for Down syndrome-associated developmental cognitive disabilities. Dev Neurosci 2011; 33:404-13. [PMID: 21865664 DOI: 10.1159/000329422] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Accepted: 03/23/2011] [Indexed: 12/19/2022] Open
Abstract
Down syndrome (DS) is mainly caused by the presence of an extra copy of human chromosome 21 (Hsa21) and is a leading genetic cause for developmental cognitive disabilities in humans. The mouse is a premier model organism for DS because the regions on Hsa21 are syntenically conserved with three regions in the mouse genome, which are located on mouse chromosome 10 (Mmu10), Mmu16 and Mmu17. With the advance of chromosomal manipulation technologies, new mouse mutants have been generated to mimic DS at both the genotypic and phenotypic levels. Further mouse-based molecular genetic studies in the future may lead to the unraveling of the mechanisms underlying DS-associated developmental cognitive disabilities, which would lay the groundwork for developing effective treatments for this phenotypic manifestation. In this review, we will discuss recent progress and future challenges in modeling DS-associated developmental cognitive disability in mice with an emphasis on hippocampus-related phenotypes.
Collapse
Affiliation(s)
- Chunhong Liu
- Children's Guild Foundation Down Syndrome Research Program and Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Gene dosage imbalance of human chromosome 21 in mouse embryonic stem cells differentiating to neurons. Gene 2011; 481:93-101. [DOI: 10.1016/j.gene.2011.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 04/15/2011] [Indexed: 01/18/2023]
|
77
|
Pennings JLA, Rodenburg W, Imholz S, Koster MPH, van Oostrom CTM, Breit TM, Schielen PCJI, de Vries A. Gene expression profiling in a mouse model identifies fetal liver- and placenta-derived potential biomarkers for Down Syndrome screening. PLoS One 2011; 6:e18866. [PMID: 21533146 PMCID: PMC3077415 DOI: 10.1371/journal.pone.0018866] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Accepted: 03/21/2011] [Indexed: 11/23/2022] Open
Abstract
Background As a first step to identify novel potential biomarkers for prenatal Down
Syndrome screening, we analyzed gene expression in embryos of wild type mice
and the Down Syndrome model Ts1Cje. Since current Down Syndrome screening
markers are derived from placenta and fetal liver, these tissues were chosen
as target. Methodology/Principal Findings Placenta and fetal liver at 15.5 days gestation were analyzed by microarray
profiling. We confirmed increased expression of genes located at the
trisomic chromosomal region. Overall, between the two genotypes more
differentially expressed genes were found in fetal liver than in placenta.
Furthermore, the fetal liver data are in line with the hematological
aberrations found in humans with Down Syndrome as well as Ts1Cje mice.
Together, we found 25 targets that are predicted (by Gene Ontology, UniProt,
or the Human Plasma Proteome project) to be detectable in human serum. Conclusions/Significance Fetal liver might harbor more promising targets for Down Syndrome screening
studies. We expect these new targets will help focus further experimental
studies on identifying and validating human maternal serum biomarkers for
Down Syndrome screening.
Collapse
Affiliation(s)
- Jeroen L A Pennings
- Laboratory for Health Protection Research (GBO), National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
78
|
Bartesaghi R, Guidi S, Ciani E. Is it possible to improve neurodevelopmental abnormalities in Down syndrome? Rev Neurosci 2011; 22:419-55. [DOI: 10.1515/rns.2011.037] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
79
|
Roubertoux PL, Carlier M. Mouse models of cognitive disabilities in trisomy 21 (Down syndrome). AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2010; 154C:400-16. [DOI: 10.1002/ajmg.c.30280] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
80
|
Costa V, Sommese L, Casamassimi A, Colicchio R, Angelini C, Marchesano V, Milone L, Farzati B, Giovane A, Fiorito C, Rienzo M, Picardi M, Avallone B, Marco Corsi M, Sarubbi B, Calabrò R, Salvatore P, Ciccodicola A, Napoli C. Impairment of circulating endothelial progenitors in Down syndrome. BMC Med Genomics 2010; 3:40. [PMID: 20836844 PMCID: PMC2949777 DOI: 10.1186/1755-8794-3-40] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Accepted: 09/13/2010] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Pathological angiogenesis represents a critical issue in the progression of many diseases. Down syndrome is postulated to be a systemic anti-angiogenesis disease model, possibly due to increased expression of anti-angiogenic regulators on chromosome 21. The aim of our study was to elucidate some features of circulating endothelial progenitor cells in the context of this syndrome. METHODS Circulating endothelial progenitors of Down syndrome affected individuals were isolated, in vitro cultured and analyzed by confocal and transmission electron microscopy. ELISA was performed to measure SDF-1α plasma levels in Down syndrome and euploid individuals. Moreover, qRT-PCR was used to quantify expression levels of CXCL12 gene and of its receptor in progenitor cells. The functional impairment of Down progenitors was evaluated through their susceptibility to hydroperoxide-induced oxidative stress with BODIPY assay and the major vulnerability to the infection with human pathogens. The differential expression of crucial genes in Down progenitor cells was evaluated by microarray analysis. RESULTS We detected a marked decrease of progenitors' number in young Down individuals compared to euploid, cell size increase and some major detrimental morphological changes. Moreover, Down syndrome patients also exhibited decreased SDF-1α plasma levels and their progenitors had a reduced expression of SDF-1α encoding gene and of its membrane receptor. We further demonstrated that their progenitor cells are more susceptible to hydroperoxide-induced oxidative stress and infection with Bartonella henselae. Further, we observed that most of the differentially expressed genes belong to angiogenesis, immune response and inflammation pathways, and that infected progenitors with trisomy 21 have a more pronounced perturbation of immune response genes than infected euploid cells. CONCLUSIONS Our data provide evidences for a reduced number and altered morphology of endothelial progenitor cells in Down syndrome, also showing the higher susceptibility to oxidative stress and to pathogen infection compared to euploid cells, thereby confirming the angiogenesis and immune response deficit observed in Down syndrome individuals.
Collapse
Affiliation(s)
- Valerio Costa
- Institute of Genetics and Biophysics A, Buzzati-Traverso, IGB-CNR, Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Abstract
A growing and interdisciplinary translational neuroscience research effort for neurodevelopmental disorders (NDDs) is investigating the mechanisms of dysfunction and testing effective treatment strategies in animal models and, when possible, in the clinic. NDDs with a genetic basis have received particular attention. Transgenic animals that mimic genetic insults responsible for disease in man have provided insight about mechanisms of dysfunction, and, surprisingly, have shown that cognitive deficits can be addressed in adult animals. This review will present recent translational research based on animal models of genetic NDDs, as well as pharmacotherapeutic strategies under development to address deficits of brain function for Down syndrome, fragile X syndrome, Rett syndrome, neurofibromatosis-1, tuberous sclerosis, and autism. Although these disorders vary in underlying causes and clinical presentation, common pathways and mechanisms for dysfunction have been observed. These include abnormal gene dosage, imbalance among neurotransmitter systems, and deficits in the development, maintenance and plasticity of neuronal circuits. NDDs affect multiple brain systems and behaviors that may be amenable to drug therapies that target distinct deficits. A primary goal of translational research is to replace symptomatic and supportive drug therapies with pharmacotherapies based on a principled understanding of the causes of dysfunction. Based on this principle, several recently developed therapeutic strategies offer clear promise for clinical development in man.
Collapse
Affiliation(s)
- Daniel Z Wetmore
- Department of Psychiatry and Behavioral Sciences, Nancy Pritzker Laboratory, Stanford University, Stanford, CA 94305-5485, USA
| | | |
Collapse
|
82
|
Abstract
Fifty years after the discovery of the etiology of Down syndrome, trisomy 21 remains the model of choice for studying human diseases resulting from the presence of a chromosome or a chromosome segment in excess. In this review, mechanisms of aneuploidy occurrence and consequences of genomic imbalances will be mainly discussed. The study of genetic markers showed that trisomy 21 results in 90% of cases from an error during maternal meiosis. Approximately 8% of cases result from an error during paternal meiosis and in 2% of cases there is a postzygotic mitotic nondisjunction. The biological basis of the effect of maternal age remains largely unknown. The absence of genetic recombination between homologous chromosomes or the presence of an exchange in telomeric position are two risk factors of non-disjunction observed in young women. Non-disjunctions associated with pericentromeric exchanges are observed with an increase in maternal age. The study of mouse models and patients with partial trisomy 21, combined with advances in knowledge of the physical map and the transcriptome, identified genes directly or indirectly involved in the pathogenesis of Down syndrome. The recent description of metabolic pathways controlled by RCAN1 and DYRK1A genes which may be involved in many biological processes and phenotypes associated with trisomy 21 allows to consider new therapeutic strategies.
Collapse
Affiliation(s)
- Catherine Turleau
- Service de cytogénétique, Hôpital Necker-Enfants malades, 149, rue de Sèvres, 75743 Paris, France
| | | |
Collapse
|
83
|
Ohnologs in the human genome are dosage balanced and frequently associated with disease. Proc Natl Acad Sci U S A 2010; 107:9270-4. [PMID: 20439718 DOI: 10.1073/pnas.0914697107] [Citation(s) in RCA: 200] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
About 30% of protein-coding genes in the human genome are related through two whole genome duplication (WGD) events. Although WGD is often credited with great evolutionary importance, the processes governing the retention of these genes and their biological significance remain unclear. One increasingly popular hypothesis is that dosage balance constraints are a major determinant of duplicate gene retention. We test this hypothesis and show that WGD-duplicated genes (ohnologs) have rarely experienced subsequent small-scale duplication (SSD) and are also refractory to copy number variation (CNV) in human populations and are thus likely to be sensitive to relative quantities (i.e., they are dosage-balanced). By contrast, genes that have experienced SSD in the vertebrate lineage are more likely to also display CNV. This supports the hypothesis of biased retention of dosage-balanced genes after WGD. We also show that ohnologs have a strong association with human disease. In particular, Down Syndrome (DS) caused by trisomy 21 is widely assumed to be caused by dosage effects, and 75% of previously reported candidate genes for this syndrome are ohnologs that experienced no other copy number changes. We propose the remaining dosage-balanced ohnologs on chromosome 21 as candidate DS genes. These observations clearly show a persistent resistance to dose changes in genes duplicated by WGD. Dosage balance constraints simultaneously explain duplicate gene retention and essentiality after WGD.
Collapse
|
84
|
Rachidi M, Lopes C. Molecular and cellular mechanisms elucidating neurocognitive basis of functional impairments associated with intellectual disability in Down syndrome. AMERICAN JOURNAL ON INTELLECTUAL AND DEVELOPMENTAL DISABILITIES 2010; 115:83-112. [PMID: 20441388 DOI: 10.1352/1944-7558-115.2.83] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2008] [Accepted: 11/05/2009] [Indexed: 05/29/2023]
Abstract
Down syndrome, the most common genetic cause of intellectual disability, is associated with brain disorders due to chromosome 21 gene overdosage. Molecular and cellular mechanisms involved in the neuromorphological alterations and cognitive impairments are reported herein in a global model. Recent advances in Down syndrome research have lead to the identification of altered molecular pathways involved in intellectual disability, such as Calcineurin/NFATs pathways, that are of crucial importance in understanding the molecular basis of intellectual disability pathogenesis in this syndrome. Potential treatments in mouse models of Down syndrome, including antagonists of NMDA or GABA(A) receptors, and microRNAs provide new avenues to develop treatments of intellectual disability. Nevertheless, understanding the links between molecular pathways and treatment strategies in human beings requires further research.
Collapse
Affiliation(s)
- Mohammed Rachidi
- University of Paris, Denis Diderot Laboratory of Genetic Dysregulation Models: Trisomy 21 and Hyperhomocysteinemia. Tour 54, Paris, France.
| | | |
Collapse
|
85
|
Abstract
Aneuploidy involves changes in chromosomal copy number compared with normal euploid genotypes. Studies of gene expression in aneuploids in a variety of species have claimed many different types of responses. Studies of individual genes suggest that there are both structural gene dosage effects and compensation in aneuploids, and that subtle trans-acting effects across the genome are quite prevalent. A discussion is presented concerning the normalization procedures for studying gene expression in aneuploids. A careful documentation of the modulations of gene expression in aneuploids should provide insight into the nature of cancerous cells and the basis of aneuploid syndromes.
Collapse
|
86
|
Makarevitch I, Harris C. Aneuploidy causes tissue-specific qualitative changes in global gene expression patterns in maize. PLANT PHYSIOLOGY 2010; 152:927-38. [PMID: 20018594 PMCID: PMC2815861 DOI: 10.1104/pp.109.150466] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Segmental aneuploidy refers to the relative excess or deficiency of specific chromosome regions. This condition results in gene dosage imbalance and often causes severe phenotypic alterations in plants and animals. The mechanisms by which gene dosage imbalance affects gene expression and phenotype are not completely clear. The effects of aneuploidy on the transcriptome may depend on the types of cells analyzed and on the developmental stage. We performed global gene expression profiling to determine the effects of segmental aneuploidy on gene expression levels in two different maize (Zea mays) tissues and a detailed analysis of expression of 30 genes affected by aneuploidy in multiple maize tissues. Different maize tissues varied in the frequency at which genes located outside of the aneuploid regions are positively or negatively regulated as well as in the degree of gene dosage compensation. Multiple genes demonstrated qualitative changes in gene expression due to aneuploidy, when the gene became ectopically expressed or completely silenced in aneuploids relative to wild-type plants. Our data strongly suggested that quantitative changes in gene expression at developmental transition points caused by variation in gene copy number progressed through tissue development and resulted in stable qualitative changes in gene expression patterns. Thus, aneuploidy in maize results in alterations of gene expression patterns that differ between tissues and developmental stages of maize seedlings.
Collapse
Affiliation(s)
- Irina Makarevitch
- Biology Department, Hamline University, Saint Paul, Minnesota 55104, USA.
| | | |
Collapse
|
87
|
Cramer NP, Best TK, Stoffel M, Siarey RJ, Galdzicki Z. GABAB–GIRK2-Mediated Signaling in Down Syndrome. GABABRECEPTOR PHARMACOLOGY - A TRIBUTE TO NORMAN BOWERY 2010; 58:397-426. [DOI: 10.1016/s1054-3589(10)58015-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
88
|
Reinholdt LG, Czechanski A, Kamdar S, King BL, Sun F, Handel MA. Meiotic behavior of aneuploid chromatin in mouse models of Down syndrome. Chromosoma 2009; 118:723-36. [PMID: 19639331 PMCID: PMC2848991 DOI: 10.1007/s00412-009-0230-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Revised: 07/07/2009] [Accepted: 07/08/2009] [Indexed: 12/23/2022]
Abstract
Aneuploidy, which leads to unpaired chromosomal axes during meiosis, is frequently accompanied by infertility. We previously showed, using three mouse models of Down syndrome, that it is an extra chromosome, but not extra gene dose, that is associated with male infertility and virtual absence of post-meiotic gem cells. Here, we test the hypothesis that aneuploid segments are differentially modified and expressed during meiosis, depending on whether they are present as an extra chromosome or not. In all three models examined, the trisomic region lacks a pairing partner, but in one case, spermatocytes have an extra (and unpaired) chromosome, while the two other models involve translocation of the trisomic region rather than an extra chromosome. An extra unpaired chromosome was always modified by phosphorylation of histone H2AX and lacked RNA PolII. But in the case of trisomic regions attached to a paired chromosome, assembly of these protein modifications was affected by the position of a trisomic region relative to a centromere and the physical extent of the unpaired chromatin. Analysis of gene expression in testes revealed that extra copy number alone was not sufficient for meiotic upregulation of genes in the trisomic interval. Additionally and unexpectedly, presence of meiotic gene silencing chromatin modifications was not sufficient for downregulation of genes in unpaired trisomic chromatin. Thus, the meiotic chromatin modifications that are cytologically visible are unlikely to be directly involved in sterility versus fertility of DS models. Finally, the presence of an extra unpaired chromosome, but not the presence of extra (trisomic) genes, caused global deregulation of transcription in spermatocytes. These results reveal mechanisms by which an extra chromosome, but not trisomic gene dose, impact on meiotic progress and infertility.
Collapse
|
89
|
Hu Y, Lehrach H, Janitz M. Apoptosis screening of human chromosome 21 proteins reveals novel cell death regulators. Mol Biol Rep 2009; 37:3381-7. [PMID: 20091128 PMCID: PMC2941049 DOI: 10.1007/s11033-009-9926-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2009] [Accepted: 11/16/2009] [Indexed: 11/28/2022]
Abstract
The functional analysis of chromosome 21 (Chr21) proteins is of great medical relevance. This refers, in particular, to the trisomy of human Chr21, which results in Down’s syndrome, a complex developmental and neurodegenerative disease. In a previous study we analyzed 89 human Chr21 genes for the subcellular localization of their encoded proteins using a transfected-cell array technique. In the present study, the results of the follow-up investigation are presented in which 52 human Chr21 genes were over-expressed in HEK cells using the transfected-cell array platform, and the effect of this protein over-expression on the induction of apoptosis has been analyzed. We found that the over-expression of two Chr21 proteins (claudin-14 and -8) induced cell death independent of the classic caspase-mediated apoptosis. Our results strongly suggest the functional involvement of claudins in the control of the cell cycle and regulation of the cell death induction mechanism.
Collapse
Affiliation(s)
- Yuhui Hu
- Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, Ihnestr. 73, 14195, Berlin, Germany
| | | | | |
Collapse
|
90
|
Pennings JLA, Koster MPH, Rodenburg W, Schielen PCJI, de Vries A. Discovery of novel serum biomarkers for prenatal Down syndrome screening by integrative data mining. PLoS One 2009; 4:e8010. [PMID: 19956656 PMCID: PMC2777317 DOI: 10.1371/journal.pone.0008010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Accepted: 11/03/2009] [Indexed: 12/23/2022] Open
Abstract
Background To facilitate the experimental search for novel maternal serum biomarkers in prenatal Down Syndrome screening, we aimed to create a set of candidate biomarkers using a data mining approach. Methodology/Principal Findings Because current screening markers are derived from either fetal liver or placental trophoblasts, we reasoned that new biomarkers can primarily be found to be derived from these two tissues. By applying a three-stage filtering strategy on publicly available data from different sources, we identified 49 potential blood-detectable protein biomarkers. Our set contains three biomarkers that are currently widely used in either first- or second-trimester screening (AFP, PAPP-A and fβ-hCG), as well as ten other proteins that are or have been examined as prenatal serum markers. This supports the effectiveness of our strategy and indicates the set contains other markers potentially applicable for screening. Conclusions/Significance We anticipate the set will help support further experimental studies for the identification of new Down Syndrome screening markers in maternal blood.
Collapse
Affiliation(s)
- Jeroen L A Pennings
- Laboratory for Health Protection Research (GBO), National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands.
| | | | | | | | | |
Collapse
|
91
|
Moldrich RX, Dauphinot L, Laffaire J, Vitalis T, Hérault Y, Beart PM, Rossier J, Vivien D, Gehrig C, Antonarakis SE, Lyle R, Potier MC. Proliferation deficits and gene expression dysregulation in Down's syndrome (Ts1Cje) neural progenitor cells cultured from neurospheres. J Neurosci Res 2009; 87:3143-52. [PMID: 19472221 DOI: 10.1002/jnr.22131] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Down's syndrome neurophenotypes are characterized by mental retardation and a decreased brain volume. To identify whether deficits in proliferation could be responsible for this phenotype, neural progenitor cells were isolated from the developing E14 neocortex of Down's syndrome partial trisomy Ts1Cje mice and euploid (WT) littermates and grown as neurospheres. Ts1Cje neural progenitors proliferated at a slower rate, because of a longer cell cycle, and a greater number of cells were positive for glial fibrillary acidic protein. An increase in cell death was also noted. Gene expression profiles of neural progenitor cells from Ts1Cje and WT showed that 54% of triploid genes had expression ratios (Ts1Cje/WT) significantly greater than the expected diploid gene ratio of 1.0. Some diploid genes associated with proliferation, differentiation, and glial function were dysregulated. Interestingly, proliferation and gene expression dysregulation detected in the Ts1Cje mice did not require overexpression of the chromosome 21 genes amyloid precursor protein (App) and soluble superoxide dismutase 1 (Sod1).
Collapse
Affiliation(s)
- Randal X Moldrich
- Laboratoire de Neurobiologie et Diversité Cellulaire, CNRS UMR7637, ESPCI, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Pereira PL, Magnol L, Sahún I, Brault V, Duchon A, Prandini P, Gruart A, Bizot JC, Chadefaux-Vekemans B, Deutsch S, Trovero F, Delgado-García JM, Antonarakis SE, Dierssen M, Herault Y. 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 2009; 18:4756-69. [PMID: 19783846 PMCID: PMC2778371 DOI: 10.1093/hmg/ddp438] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Mental retardation in Down syndrome (DS), the most frequent trisomy in humans, varies from moderate to severe. Several studies both in human and based on mouse models identified some regions of human chromosome 21 (Hsa21) as linked to cognitive deficits. However, other intervals such as the telomeric region of Hsa21 may contribute to the DS phenotype but their role has not yet been investigated in detail. Here we show that the trisomy of the 12 genes, found in the 0.59 Mb (Abcg1–U2af1) Hsa21 sub-telomeric region, in mice (Ts1Yah) produced defects in novel object recognition, open-field and Y-maze tests, similar to other DS models, but induces an improvement of the hippocampal-dependent spatial memory in the Morris water maze along with enhanced and longer lasting long-term potentiation in vivo in the hippocampus. Overall, we demonstrate the contribution of the Abcg1–U2af1 genetic region to cognitive defect in working and short-term recognition memory in DS models. Increase in copy number of the Abcg1–U2af1 interval leads to an unexpected gain of cognitive function in spatial learning. Expression analysis pinpoints several genes, such as Ndufv3, Wdr4, Pknox1 and Cbs, as candidates whose overexpression in the hippocampus might facilitate learning and memory in Ts1Yah mice. Our work unravels the complexity of combinatorial genetic code modulating different aspect of mental retardation in DS patients. It establishes definitely the contribution of the Abcg1–U2af1 orthologous region to the DS etiology and suggests new modulatory pathways for learning and memory.
Collapse
Affiliation(s)
- Patricia Lopes Pereira
- Molecular Embryology and Immunology, Université d'Orléans, UMR6218, Orléans Cedex 2, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Choi JHK, Berger JD, Mazzella MJ, Morales-Corraliza J, Cataldo AM, Nixon RA, Ginsberg SD, Levy E, Mathews PM. Age-dependent dysregulation of brain amyloid precursor protein in the Ts65Dn Down syndrome mouse model. J Neurochem 2009; 110:1818-27. [PMID: 19619138 PMCID: PMC2744432 DOI: 10.1111/j.1471-4159.2009.06277.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Individuals with Down syndrome develop beta-amyloid deposition characteristic of early-onset Alzheimer's disease (AD) in mid-life, presumably because of an extra copy of the chromosome 21-located amyloid precursor protein (App) gene. App mRNA and APP metabolite levels were assessed in the brains of Ts65Dn mice, a mouse model of Down syndrome, using quantitative PCR, western blot analysis, immunoprecipitation, and ELISAs. In spite of the additional App gene copy, App mRNA, APP holoprotein, and all APP metabolite levels in the brains of 4-month-old trisomic mice were not increased compared with the levels seen in diploid littermate controls. However starting at 10 months of age, brain APP levels were increased proportional to the App gene dosage imbalance reflecting increased App message levels in Ts65Dn mice. Similar to APP levels, soluble amino-terminal fragments of APP (sAPPalpha and sAPPbeta) were increased in Ts65Dn mice compared with diploid mice at 12 months but not at 4 months of age. Brain levels of both Abeta40 and Abeta42 were not increased in Ts65Dn mice compared with diploid mice at all ages examined. Therefore, multiple mechanisms contribute to the regulation towards diploid levels of APP metabolites in the Ts65Dn mouse brain.
Collapse
Affiliation(s)
- Jennifer H K Choi
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York 10962, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Dierssen M, Herault Y, Estivill X. Aneuploidy: from a physiological mechanism of variance to Down syndrome. Physiol Rev 2009; 89:887-920. [PMID: 19584316 DOI: 10.1152/physrev.00032.2007] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Quantitative differences in gene expression emerge as a significant source of variation in natural populations, representing an important substrate for evolution and accounting for a considerable fraction of phenotypic diversity. However, perturbation of gene expression is also the main factor in determining the molecular pathogenesis of numerous aneuploid disorders. In this review, we focus on Down syndrome (DS) as the prototype of "genomic disorder" induced by copy number change. The understanding of the pathogenicity of the extra genomic material in trisomy 21 has accelerated in the last years due to the recent advances in genome sequencing, comparative genome analysis, functional genome exploration, and the use of model organisms. We present recent data on the role of genome-altering processes in the generation of diversity in DS neural phenotypes focusing on the impact of trisomy on brain structure and mental retardation and on biological pathways and cell types in target brain regions (including prefrontal cortex, hippocampus, cerebellum, and basal ganglia). We also review the potential that genetically engineered mouse models of DS bring into the understanding of the molecular biology of human learning disorders.
Collapse
Affiliation(s)
- Mara Dierssen
- Genes and Disease Program, Genomic Regulation Center-CRG, Pompeu Fabra University, Barcelona Biomedical Research Park, Dr Aiguader 88, PRBB building E, Barcelona 08003, Catalonia, Spain.
| | | | | |
Collapse
|
95
|
Enhanced apoptosis during early neuronal differentiation in mouse ES cells with autosomal imbalance. Cell Res 2009; 19:247-58. [PMID: 19015669 DOI: 10.1038/cr.2008.305] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Although particular chromosomal syndromes are phenotypically and clinically distinct, the majority of individuals with autosomal imbalance, such as aneuploidy, manifest mental retardation. A common abnormal phenotype of Down syndrome (DS), the most prevalent autosomal aneuploidy, shows a reduction in both the number and the density of neurons in the brain. As a DS model, we have recently created chimeric mice from ES cells containing a single human chromosome 21. The mice mimicked the characteristic phenotypic features of DS, and ES cells showed a higher incidence of apoptosis during early neuronal differentiation in vitro. In this study, we examined the induction of anomalous early neural development by aneuploidy in mouse ES cells by transferring various human chromosomes or additional mouse chromosomes. Results showed an elevated incidence of apoptosis in all autosome-aneuploid clones examined during early neuronal differentiation in vitro. Further, cDNA microarray analysis revealed a common cluster of down-regulated genes, of which eight known genes are related to cell proliferation, neurite outgrowth and differentiation. Importantly, targeting of these genes by siRNA knockdown in normal mouse ES cells led to enhanced apoptosis during early neuronal differentiation. These findings strongly suggest that autosomal imbalance is associated with general neuronal loss through a common molecular mechanism for apoptosis.
Collapse
|
96
|
Rachidi M, Delezoide AL, Delabar JM, Lopes C. A quantitative assessment of gene expression (QAGE) reveals differential overexpression of DOPEY2, a candidate gene for mental retardation, in Down syndrome brain regions. Int J Dev Neurosci 2009; 27:393-8. [PMID: 19460634 DOI: 10.1016/j.ijdevneu.2009.02.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2008] [Revised: 01/30/2009] [Accepted: 02/03/2009] [Indexed: 11/25/2022] Open
Abstract
The brain alterations and mental retardation in Down syndrome are associated with overdosage of chromosome 21 genes. To shed light on the understanding of the molecular effect of this genetic overdosage, gene expression studies have crucial importance to quantify expression variations in Down syndrome tissues compared to normal ones. Herein, an in situ Quantitative Assessment of Gene Expression (QAGE) was used to quantify and statistically analyze, for the first time, DOPEY2 expression variations in different regions of the Down syndrome human fetal brains and to compare them to corresponding normal brains. DOPEY2, which is localized in the Down Syndrome Critical Region (DSCR) and is a candidate gene for neurological alterations in Down syndrome, showed a delimited regional and cellular expression pattern in the cortex, hippocampus and cerebellum, characterized by different transcriptional intensities in both normal and trisomic brains. DOPEY2 is overexpressed more than 50% (1.79-, 1.97- and 2.12-folds in the cortex, cerebellum and hippocampus, respectively), and showed statistically significant differences in the overexpression ratios in the three brain regions expressing DOPEY2. The demonstration of differential DOPEY2 expression and overexpression in human fetal brains suggests that this gene is submitted to a complex transcriptional control and could depend from other human chromosome 21 genes. Moreover, DOPEY2 overexpression in the brain regions, that are altered in Down syndrome patients and involved in learning and memory processes, is in agreement to the hypothesis that this gene plays a potential role in functional brain alterations and in the pathogenesis of mental retardation in Down syndrome. This new in situ QAGE approach allowed quantitative measurements of transcriptional changes and statistical evaluations of the expression and overexpression patterns of DOPEY2 at specific regions of the brain, which is a complementary approach to qRT-PCR and microarray for transcriptome study. Moreover, this approach could be a powerful tool to study the candidate chromosome 21 genes for Down syndrome and other pathologies caused by regionalized quantitative transcriptional alterations, for greater interpretation of functional processes driving gene expression.
Collapse
Affiliation(s)
- Mohammed Rachidi
- Laboratory of Genetic Dysregulation Models: Trisomy 21 and Hyperhomocysteinemia, EA 3508, University Paris 7-Denis Diderot, Paris, France.
| | | | | | | |
Collapse
|
97
|
Arnold AP, Itoh Y, Melamed E. A bird's-eye view of sex chromosome dosage compensation. Annu Rev Genomics Hum Genet 2008; 9:109-27. [PMID: 18489256 DOI: 10.1146/annurev.genom.9.081307.164220] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Intensive study of a few genetically tractable species with XX/XY sex chromosomes has produced generalizations about the process of sex chromosome dosage compensation that do not fare well when applied to ZZ/ZW sex chromosome systems, such as those in birds. The inherent sexual imbalance in dose of sex chromosome genes has led to the evolution of sex-chromosome-wide mechanisms for balancing gene dosage between the sexes and relative to autosomal genes. Recent advances in our knowledge of avian genomes have led to a reexamination of sex-specific dosage compensation (SSDC) in birds, which is less effective than in known XX/XY systems. Insights about the mechanisms of SSDC in birds also suggest similarities to and differences from those in XX/XY species. Birds are thus offering new opportunities for studying dosage compensation in a ZZ/ZW system, which should shed light on the evolution of SSDC more broadly.
Collapse
Affiliation(s)
- Arthur P Arnold
- Department of Physiological Science and Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, California 90095, USA.
| | | | | |
Collapse
|
98
|
Lyle R, Béna F, Gagos S, Gehrig C, Lopez G, Schinzel A, Lespinasse J, Bottani A, Dahoun S, Taine L, Doco-Fenzy M, Cornillet-Lefèbvre P, Pelet A, Lyonnet S, Toutain A, Colleaux L, Horst J, Kennerknecht I, Wakamatsu N, Descartes M, Franklin JC, Florentin-Arar L, Kitsiou S, Aït Yahya-Graison E, Costantine M, Sinet PM, Delabar JM, Antonarakis SE. 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 2008; 17:454-66. [PMID: 19002211 DOI: 10.1038/ejhg.2008.214] [Citation(s) in RCA: 209] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Down syndrome (DS) is one of the most frequent congenital birth defects, and the most common genetic cause of mental retardation. In most cases, DS results from the presence of an extra copy of chromosome 21. DS has a complex phenotype, and a major goal of DS research is to identify genotype-phenotype correlations. Cases of partial trisomy 21 and other HSA21 rearrangements associated with DS features could identify genomic regions associated with specific phenotypes. We have developed a BAC array spanning HSA21q and used array comparative genome hybridization (aCGH) to enable high-resolution mapping of pathogenic partial aneuploidies and unbalanced translocations involving HSA21. We report the identification and mapping of 30 pathogenic chromosomal aberrations of HSA21 consisting of 19 partial trisomies and 11 partial monosomies for different segments of HSA21. The breakpoints have been mapped to within approximately 85 kb. The majority of the breakpoints (26 of 30) for the partial aneuploidies map within a 10-Mb region. Our data argue against a single DS critical region. We identify susceptibility regions for 25 phenotypes for DS and 27 regions for monosomy 21. However, most of these regions are still broad, and more cases are needed to narrow down the phenotypic maps to a reasonable number of candidate genomic elements per phenotype.
Collapse
Affiliation(s)
- Robert Lyle
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Duffy KJ, Littrell J, Locke A, Sherman SL, Olivier M. A novel procedure for genotyping of single nucleotide polymorphisms in trisomy with genomic DNA and the invader assay. Nucleic Acids Res 2008; 36:e145. [PMID: 18940863 PMCID: PMC2602776 DOI: 10.1093/nar/gkn736] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Individuals with trisomy 21 display complex phenotypes with differing degrees of severity. Numerous reliable methods have been established to diagnose the initial trisomy in these patients, but the identification and characterization of the genetic basis of the phenotypic variation in individuals with trisomy remains challenging. To date, methods that can accurately determine genotypes in trisomic DNA samples are expensive, require specialized equipment and complicated analyses. Here we report proof-of-concept results for an Invader® assay-based genotyping procedure that can determine SNP genotypes in trisomic genomic DNA samples in a simple and cost-effective manner. The procedure requires only two experimental steps: a real-time measurement of the fluorescent Invader® signal and analysis with a specifically designed clustering algorithm. The approach was tested using genomic DNA samples from 23 individuals with trisomy 21, and results were compared to genotypes previously determined with pyrosequencing. Additional assays for 15 SNPs were tested in a set of 21 DNA samples to assess assay performance. Our method successfully identified the correct SNP genotypes for the trisomic genomic DNA samples tested, and thus provides an alternative to determine SNP genotypes in trisomic DNA samples for subsequent association studies in patients with Down syndrome and other trisomies.
Collapse
Affiliation(s)
- Kelly J Duffy
- Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | | | | | | |
Collapse
|
100
|
Canzonetta C, Mulligan C, Deutsch S, Ruf S, O'Doherty A, Lyle R, Borel C, Lin-Marq N, Delom F, Groet J, Schnappauf F, De Vita S, Averill S, Priestley JV, Martin JE, Shipley J, Denyer G, Epstein CJ, Fillat C, Estivill X, Tybulewicz VL, Fisher EM, Antonarakis SE, Nizetic D. DYRK1A-dosage imbalance perturbs NRSF/REST levels, deregulating pluripotency and embryonic stem cell fate in Down syndrome. Am J Hum Genet 2008; 83:388-400. [PMID: 18771760 PMCID: PMC2556438 DOI: 10.1016/j.ajhg.2008.08.012] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Revised: 07/10/2008] [Accepted: 08/20/2008] [Indexed: 10/21/2022] Open
Abstract
Down syndrome (DS) is the most common cause of mental retardation. Many neural phenotypes are shared between DS individuals and DS mouse models; however, the common underlying molecular pathogenetic mechanisms remain unclear. Using a transchromosomic model of DS, we show that a 30%-60% reduced expression of Nrsf/Rest (a key regulator of pluripotency and neuronal differentiation) is an alteration that persists in trisomy 21 from undifferentiated embryonic stem (ES) cells to adult brain and is reproducible across several DS models. Using partially trisomic ES cells, we map this effect to a three-gene segment of HSA21, containing DYRK1A. We independently identify the same locus as the most significant eQTL controlling REST expression in the human genome. We show that specifically silencing the third copy of DYRK1A rescues Rest levels, and we demonstrate altered Rest expression in response to inhibition of DYRK1A expression or kinase activity, and in a transgenic Dyrk1A mouse. We reveal that undifferentiated trisomy 21 ES cells show DYRK1A-dose-sensitive reductions in levels of some pluripotency regulators, causing premature expression of transcription factors driving early endodermal and mesodermal differentiation, partially overlapping recently reported downstream effects of Rest +/-. They produce embryoid bodies with elevated levels of the primitive endoderm progenitor marker Gata4 and a strongly reduced neuroectodermal progenitor compartment. Our results suggest that DYRK1A-mediated deregulation of REST is a very early pathological consequence of trisomy 21 with potential to disturb the development of all embryonic lineages, warranting closer research into its contribution to DS pathology and new rationales for therapeutic approaches.
Collapse
Affiliation(s)
- Claudia Canzonetta
- Institute of Cell and Molecular Science, Barts & The London, Queen Mary's School of Medicine and Dentistry, University of London, 4 Newark Street, London E1 2AT, UK
| | - Claire Mulligan
- Institute of Cell and Molecular Science, Barts & The London, Queen Mary's School of Medicine and Dentistry, University of London, 4 Newark Street, London E1 2AT, UK
| | - Samuel Deutsch
- Department of Genetic Medicine and Development, Geneva University Medical School, Geneva CH-1211, Switzerland
| | - Sandra Ruf
- National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
| | - Aideen O'Doherty
- National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Robert Lyle
- Department of Genetic Medicine and Development, Geneva University Medical School, Geneva CH-1211, Switzerland
| | - Christelle Borel
- Department of Genetic Medicine and Development, Geneva University Medical School, Geneva CH-1211, Switzerland
| | - Nathalie Lin-Marq
- Department of Genetic Medicine and Development, Geneva University Medical School, Geneva CH-1211, Switzerland
| | - Frederic Delom
- Institute of Cell and Molecular Science, Barts & The London, Queen Mary's School of Medicine and Dentistry, University of London, 4 Newark Street, London E1 2AT, UK
| | - Jürgen Groet
- Institute of Cell and Molecular Science, Barts & The London, Queen Mary's School of Medicine and Dentistry, University of London, 4 Newark Street, London E1 2AT, UK
| | - Felix Schnappauf
- Institute of Cell and Molecular Science, Barts & The London, Queen Mary's School of Medicine and Dentistry, University of London, 4 Newark Street, London E1 2AT, UK
| | - Serena De Vita
- Institute of Cell and Molecular Science, Barts & The London, Queen Mary's School of Medicine and Dentistry, University of London, 4 Newark Street, London E1 2AT, UK
| | - Sharon Averill
- Institute of Cell and Molecular Science, Barts & The London, Queen Mary's School of Medicine and Dentistry, University of London, 4 Newark Street, London E1 2AT, UK
| | - John V. Priestley
- Institute of Cell and Molecular Science, Barts & The London, Queen Mary's School of Medicine and Dentistry, University of London, 4 Newark Street, London E1 2AT, UK
| | - Joanne E. Martin
- Institute of Cell and Molecular Science, Barts & The London, Queen Mary's School of Medicine and Dentistry, University of London, 4 Newark Street, London E1 2AT, UK
| | - Janet Shipley
- The Institute of Cancer Research, Sutton, Surrey SM2 5NG, UK
| | - Gareth Denyer
- Department of Biochemistry, University of Sydney, Sydney NSW 2006, Australia
| | - Charles J. Epstein
- Department of Pediatrics, University of California, San Francisco, CA 94143-2911, USA
| | - Cristina Fillat
- Genes and Disease Program, Center for Genomic Regulation (CRG-UPF), and CIBERESP and CIBERER, Barcelona 08003, Spain
| | - Xavier Estivill
- Genes and Disease Program, Center for Genomic Regulation (CRG-UPF), and CIBERESP and CIBERER, Barcelona 08003, Spain
| | | | - Elizabeth M.C. Fisher
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Stylianos E. Antonarakis
- Department of Genetic Medicine and Development, Geneva University Medical School, Geneva CH-1211, Switzerland
| | - Dean Nizetic
- Institute of Cell and Molecular Science, Barts & The London, Queen Mary's School of Medicine and Dentistry, University of London, 4 Newark Street, London E1 2AT, UK
| |
Collapse
|