Investigation of post-transcriptional gene regulatory networks associated with autism spectrum disorders by microRNA expression profiling of lymphoblastoid cell lines

Tewarit Sarachana, Rulun Zhou, Guang Chen, Husseini K Manji, Valerie W Hu, Tewarit Sarachana, Rulun Zhou, Guang Chen, Husseini K Manji, Valerie W Hu

Abstract

Background: Autism spectrum disorders (ASD) are neurodevelopmental disorders characterized by abnormalities in reciprocal social interactions and language development and/or usage, and by restricted interests and repetitive behaviors. Differential gene expression of neurologically relevant genes in lymphoblastoid cell lines from monozygotic twins discordant in diagnosis or severity of autism suggested that epigenetic factors such as DNA methylation or microRNAs (miRNAs) may be involved in ASD.

Methods: Global miRNA expression profiling using lymphoblasts derived from these autistic twins and unaffected sibling controls was therefore performed using high-throughput miRNA microarray analysis. Selected differentially expressed miRNAs were confirmed by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) analysis, and the putative target genes of two of the confirmed miRNA were validated by knockdown and overexpression of the respective miRNAs.

Results: Differentially expressed miRNAs were found to target genes highly involved in neurological functions and disorders in addition to genes involved in gastrointestinal diseases, circadian rhythm signaling, as well as steroid hormone metabolism and receptor signaling. Novel network analyses of the putative target genes that were inversely expressed relative to the relevant miRNA in these same samples further revealed an association with ASD and other co-morbid disorders, including muscle and gastrointestinal diseases, as well as with biological functions implicated in ASD, such as memory and synaptic plasticity. Putative gene targets (ID3 and PLK2) of two RT-PCR-confirmed brain-specific miRNAs (hsa-miR-29b and hsa-miR-219-5p) were validated by miRNA overexpression or knockdown assays, respectively. Comparisons of these mRNA and miRNA expression levels between discordant twins and between case-control sib pairs show an inverse relationship, further suggesting that ID3 and PLK2 are in vivo targets of the respective miRNA. Interestingly, the up-regulation of miR-23a and down-regulation of miR-106b in this study reflected miRNA changes previously reported in post-mortem autistic cerebellum by Abu-Elneel et al. in 2008. This finding validates these differentially expressed miRNAs in neurological tissue from a different cohort as well as supports the use of the lymphoblasts as a surrogate to study miRNA expression in ASD.

Conclusions: Findings from this study strongly suggest that dysregulation of miRNA expression contributes to the observed alterations in gene expression and, in turn, may lead to the pathophysiological conditions underlying autism.

Figures

Figure 1
Figure 1
Schematic flow diagram describing procedures used to identify inversely correlated differentially expressed putatitve target genes of the differentially expressed miRNAs. Tens of thousands of putative target genes are associated with the 43 differentially expressed miRNAs, some of which are overlapping between different miRNAs. For the correlation analyses, we used all of the putative target genes.
Figure 2
Figure 2
Hierarchical cluster analysis and principal component analysis of significantly differentially expressed miRNAs from the Pavlidis template matching analysis. (a) Unsupervised hierarchical cluster analysis of 43 significantly differentially expressed miRNAs between all autistic individuals (red bar) and controls (turquoise bar) shows the distinct miRNA expression pattern of the two groups (P < 0.05). The individual samples are coded as follows: AT, autistic twin; AS, autistic sibling; CT, control, undiagnosed twin; CS, control, nonautistic sibling; C_6a/b, nonautistic, monozygotic twins a and b. The same numbers following the sample descriptors indicate members of the same family. (b) Principal component analysis of the samples based on the same set of miRNAs reduces the dimensionality of the data and shows the clear separation between the autistic individuals (red) and the controls (turquoise).
Figure 3
Figure 3
Results of TaqMan miRNA qRT-PCR analyses of four brain-associated miRNAs (hsa-miR-219-5p, hsa-miR-139-5p, hsa-miR-29b, and hsa-miR-103) in autistic and control lymphoblastoid cell lines. Expression levels of selected miRNAs associated with brain development from TaqMan qRT-PCR analyses confirm data obtained by miRNA microarrays. Green bars, qRT-PCR data; orange bars, DNA microarray data. Error bars represent standard errors associated with miRNA Taqman qRT-PCR or miRNA microarray analyses (hsa-miR-219-5p/hsa-miR-29b/hsa-miR-103, n = 5 case-control pairs; hsa-miR-139-5p, n = 4 pairs).
Figure 4
Figure 4
Relationships between differentially expressed miRNAs, putative target genes, and functions. Network and pathway analysis using Pathway Studio 5 shows the relationships among the significantly differentially expressed miRNAs, potential target genes (expression cutoff log2 ratio ≥± 0.4), and biological functions and disorders implicated by the differentially expressed target genes. Up-regulated genes and miRNAs are in red; down-regulated genes and miRNAs are in green.
Figure 5
Figure 5
Validation of miRNA targets. Three LCLs from non-autistic individuals were transfected with hsa-miR-29b pre-miR precursor, hsa-miR-219b anti-miR inhibitor, pre-miR negative control, or anti-miR negative control. At 72 hours after transfection, qRT-PCR analyses were conducted to determine expression of PLK2 and ID3 genes in the pre-miR/anti-miR-transfected LCLs (red), compared to respective pre-miR/anti-miR negative controls (navy). (a, b) Expression of PLK2 was significantly increased in the LCLs transfected with anti-miR-219-5p (a), whereas ID3 expression was significantly decreased in pre-miR-29b-transfected LCLs (b). The error bars show the standard error among the technical replicates. *P < 0.05.

References

    1. Volkmar F, Chawarska K, Klin A. Autism in infancy and early childhood. Annu Rev Psychol. 2005;56:315–336. doi: 10.1146/annurev.psych.56.091103.070159.
    1. Barton M, Volkmar F. How commonly are known medical conditions associated with autism? J Autism Dev Disord. 1998;28:273–278. doi: 10.1023/A:1026052417561.
    1. Rutter M, Bailey A, Bolton P, Le Couteur A. Autism and known medical conditions: myth and substance. J Child Psychol Psychiatry. 1994;35:311–322. doi: 10.1111/j.1469-7610.1994.tb01164.x.
    1. Volkmar FR, Lord C, Bailey A, Schultz RT, Klin A. Autism and pervasive developmental disorders. J Child Psychol Psychiatry. 2004;45:135–170. doi: 10.1046/j.0021-9630.2003.00317.x.
    1. International Molecular Genetic Study of Autism Consortium (IMGSAC) A genomewide screen for autism: strong evidence for linkage to chromosomes 2q, 7q, and 16p. Am J Hum Genet. 2001;69:570–581. doi: 10.1086/323264.
    1. Auranen M, Vanhala R, Varilo T, Ayers K, Kempas E, Ylisaukko-Oja T, Sinsheimer JS, Peltonen L, Jarvela I. A genomewide screen for autism-spectrum disorders: evidence for a major susceptibility locus on chromosome 3q25-27. Am J Hum Genet. 2002;71:777–790. doi: 10.1086/342720.
    1. Yonan AL, Alarcon M, Cheng R, Magnusson PK, Spence SJ, Palmer AA, Grunn A, Juo SH, Terwilliger JD, Liu J, Cantor RM, Geschwind DH, Gilliam TC. A genomewide screen of 345 families for autism-susceptibility loci. Am J Hum Genet. 2003;73:886–897. doi: 10.1086/378778.
    1. Philippe A, Martinez M, Guilloud-Bataille M, Gillberg C, Rastam M, Sponheim E, Coleman M, Zappella M, Aschauer H, Van Maldergem L, Penet C, Feingold J, Brice A, Leboyer M. Genome-wide scan for autism susceptibility genes. Paris Autism Research International Sibpair Study. Hum Mol Genet. 1999;8:805–812. doi: 10.1093/hmg/8.5.805.
    1. International Molecular Genetic Study of Autism Consortium (IMGSAC) Further characterization of the autism susceptibility locus AUTS1 on chromosome 7q. Hum Mol Genet. 2001;10:973–982. doi: 10.1093/hmg/10.9.973.
    1. McCauley JL, Li C, Jiang L, Olson LM, Crockett G, Gainer K, Folstein SE, Haines JL, Sutcliffe JS. Genome-wide and Ordered-Subset linkage analyses provide support for autism loci on 17q and 19p with evidence of phenotypic and interlocus genetic correlates. BMC Med Genet. 2005;6:1. doi: 10.1186/1471-2350-6-1.
    1. Weiss LA, Shen Y, Korn JM, Arking DE, Miller DT, Fossdal R, Saemundsen E, Stefansson H, Ferreira MA, Green T, Platt OS, Ruderfer DM, Walsh CA, Altshuler D, Chakravarti A, Tanzi RE, Stefansson K, Santangelo SL, Gusella JF, Sklar P, Wu BL, Daly MJ. Autism Consortium. Association between microdeletion and microduplication at 16p11.2 and autism. N Engl J Med. 2008;358:667–675. doi: 10.1056/NEJMoa075974.
    1. Lise MF, El-Husseini A. The neuroligin and neurexin families: from structure to function at the synapse. Cell Mol Life Sci. 2006;63:1833–1849. doi: 10.1007/s00018-006-6061-3.
    1. Jamain S, Quach H, Betancur C, Rastam M, Colineaux C, Gillberg IC, Soderstrom H, Giros B, Leboyer M, Gillberg C, Bourgeron T. Paris Autism Research International Sibpair Study. Mutations of the X-linked genes encoding neuroligins NLGN3 and NLGN4 are associated with autism. Nat Genet. 2003;34:27–29. doi: 10.1038/ng1136.
    1. Durand CM, Betancur C, Boeckers TM, Bockmann J, Chaste P, Fauchereau F, Nygren G, Rastam M, Gillberg IC, Anckarsater H, Sponheim E, Goubran-Botros H, Delorme R, Chabane N, Mouren-Simeoni MC, de Mas P, Bieth E, Roge B, Heron D, Burglen L, Gillberg C, Leboyer M, Bourgeron T. Mutations in the gene encoding the synaptic scaffolding protein SHANK3 are associated with autism spectrum disorders. Nat Genet. 2007;39:25–27. doi: 10.1038/ng1933.
    1. Kim HG, Kishikawa S, Higgins AW, Seong IS, Donovan DJ, Shen Y, Lally E, Weiss LA, Najm J, Kutsche K, Descartes M, Holt L, Braddock S, Troxell R, Kaplan L, Volkmar F, Klin A, Tsatsanis K, Harris DJ, Noens I, Pauls DL, Daly MJ, MacDonald ME, Morton CC, Quade BJ, Gusella JF. Disruption of neurexin 1 associated with autism spectrum disorder. Am J Hum Genet. 2008;82:199–207. doi: 10.1016/j.ajhg.2007.09.011.
    1. Arking DE, Cutler DJ, Brune CW, Teslovich TM, West K, Ikeda M, Rea A, Guy M, Lin S, Cook EH, Chakravarti A. A common genetic variant in the neurexin superfamily member CNTNAP2 increases familial risk of autism. Am J Hum Genet. 2008;82:160–164. doi: 10.1016/j.ajhg.2007.09.015.
    1. Alarcon M, Abrahams BS, Stone JL, Duvall JA, Perederiy JV, Bomar JM, Sebat J, Wigler M, Martin CL, Ledbetter DH, Nelson SF, Cantor RM, Geschwind DH. Linkage, association, and gene-expression analyses identify CNTNAP2 as an autism-susceptibility gene. Am J Hum Genet. 2008;82:150–159. doi: 10.1016/j.ajhg.2007.09.005.
    1. Bakkaloglu B, O'Roak BJ, Louvi A, Gupta AR, Abelson JF, Morgan TM, Chawarska K, Klin A, Ercan-Sencicek AG, Stillman AA, Tanriover G, Abrahams BS, Duvall JA, Robbins EM, Geschwind DH, Biederer T, Gunel M, Lifton RP, State MW. Molecular cytogenetic analysis and resequencing of contactin associated protein-like 2 in autism spectrum disorders. Am J Hum Genet. 2008;82:165–173. doi: 10.1016/j.ajhg.2007.09.017.
    1. Bourgeron T. A synaptic trek to autism. Curr Opin Neurobiol. 2009;19:231–234. doi: 10.1016/j.conb.2009.06.003.
    1. Geschwind DH. Autism: the ups and downs of neuroligin. Biol Psychiatry. 2009;66:904–905. doi: 10.1016/j.biopsych.2009.09.022.
    1. Hu VW, Frank BC, Heine S, Lee NH, Quackenbush J. Gene expression profiling of lymphoblastoid cell lines from monozygotic twins discordant in severity of autism reveals differential regulation of neurologically relevant genes. BMC Genomics. 2006;7:118. doi: 10.1186/1471-2164-7-118.
    1. Vargas DL, Nascimbene C, Krishnan C, Zimmerman AW, Pardo CA. Neuroglial activation and neuroinflammation in the brain of patients with autism. Ann Neurol. 2005;57:67–81. doi: 10.1002/ana.20315.
    1. Ambros V. The functions of animal microRNAs. Nature. 2004;431:350–355. doi: 10.1038/nature02871.
    1. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281–297. doi: 10.1016/S0092-8674(04)00045-5.
    1. Cullen BR. Transcription and processing of human microRNA precursors. Mol Cell. 2004;16:861–865. doi: 10.1016/j.molcel.2004.12.002.
    1. Kim VN. MicroRNA biogenesis: coordinated cropping and dicing. Nat Rev Mol Cell Biol. 2005;6:376–385. doi: 10.1038/nrm1644.
    1. Zamore PD, Haley B. Ribo-gnome: the big world of small RNAs. Science. 2005;309:1519–1524. doi: 10.1126/science.1111444.
    1. Rogaev EI. Small RNAs in human brain development and disorders. Biochemistry. 2005;70:1404–1407.
    1. Kim J, Inoue K, Ishii J, Vanti WB, Voronov SV, Murchison E, Hannon G, Abeliovich A. A microRNA feedback circuit in midbrain dopamine neurons. Science. 2007;317:1220–1224. doi: 10.1126/science.1140481.
    1. Mehler MF, Mattick JS. Non-coding RNAs in the nervous system. J Physiol. 2006;575:333–341. doi: 10.1113/jphysiol.2006.113191.
    1. Kapsimali M, Kloosterman WP, de Bruijn E, Rosa F, Plasterk RH, Wilson SW. MicroRNAs show a wide diversity of expression profiles in the developing and mature central nervous system. Genome Biol. 2007;8:R173. doi: 10.1186/gb-2007-8-8-r173.
    1. Caudy AA, Myers M, Hannon GJ, Hammond SM. Fragile X-related protein and VIG associate with the RNA interference machinery. Genes Dev. 2002;16:2491–2496. doi: 10.1101/gad.1025202.
    1. Ishizuka A, Siomi MC, Siomi H. A Drosophila fragile X protein interacts with components of RNAi and ribosomal proteins. Genes Dev. 2002;16:2497–2508. doi: 10.1101/gad.1022002.
    1. Jin P, Alisch RS, Warren ST. RNA and microRNAs in fragile X mental retardation. Nat Cell Biol. 2004;6:1048–1053. doi: 10.1038/ncb1104-1048.
    1. Li Y, Lin L, Jin P. The microRNA pathway and fragile X mental retardation protein. Biochim Biophys Acta. 2008;1779:702–705.
    1. Burmistrova OA, Goltsov AY, Abramova LI, Kaleda VG, Orlova VA, Rogaev EI. MicroRNA in schizophrenia: genetic and expression analysis of miR-130b (22q11) Biochemistry. 2007;72:578–582.
    1. Mourelatos Z, Dostie J, Paushkin S, Sharma A, Charroux B, Abel L, Rappsilber J, Mann M, Dreyfuss G. miRNPs: a novel class of ribonucleoproteins containing numerous microRNAs. Genes Dev. 2002;16:720–728. doi: 10.1101/gad.974702.
    1. Talebizadeh Z, Butler MG, Theodoro MF. Feasibility and relevance of examining lymphoblastoid cell lines to study role of microRNAs in autism. Autism Res. 2008;1:240–250. doi: 10.1002/aur.33.
    1. Abu-Elneel K, Liu T, Gazzaniga FS, Nishimura Y, Wall DP, Geschwind DH, Lao K, Kosik KS. Heterogeneous dysregulation of microRNAs across the autism spectrum. Neurogenetics. 2008;9:153–161. doi: 10.1007/s10048-008-0133-5.
    1. Hu VW, Nguyen A, Kim KS, Steinberg ME, Sarachana T, Scully MA, Soldin SJ, Luu T, Lee NH. Gene expression profiling of lymphoblasts from autistic and nonaffected sib pairs: altered pathways in neuronal development and steroid biosynthesis. PLoS One. 2009;4:e5775. doi: 10.1371/journal.pone.0005775.
    1. Chen Y, Kamat V, Dougherty ER, Bittner ML, Meltzer PS, Trent JM. Ratio statistics of gene expression levels and applications to microarray data analysis. Bioinformatics. 2002;18:1207–1215. doi: 10.1093/bioinformatics/18.9.1207.
    1. Dudoit S, Gentleman RC, Quackenbush J. Open source software for the analysis of microarray data. BioTechniques. 2003. pp. 45–51.
    1. Saeed AI, Sharov V, White J, Li J, Liang W, Bhagabati N, Braisted J, Klapa M, Currier T, Thiagarajan M, Sturn A, Snuffin M, Rezantsev A, Popov D, Ryltsov A, Kostukovich E, Borisovsky I, Liu Z, Vinsavich A, Trush V, Quackenbush J. TM4: a free, open-source system for microarray data management and analysis. BioTechniques. 2003;34:374–378.
    1. Quackenbush J. Microarray data normalization and transformation. Nat Genet. 2002;32(Suppl):496–501. doi: 10.1038/ng1032.
    1. Pavlidis P, Noble WS. Analysis of strain and regional variation in gene expression in mouse brain. Genome Biol. 2001;2:RESEARCH0042. doi: 10.1186/gb-2001-2-10-research0042.
    1. Griffiths-Jones S, Grocock RJ, van Dongen S, Bateman A, Enright AJ. miRBase: microRNA sequences, targets and gene nomenclature. Nucleic Acids Res. 2006;34:D140–144. doi: 10.1093/nar/gkj112.
    1. Rehmsmeier M, Steffen P, Hochsmann M, Giegerich R. Fast and effective prediction of microRNA/target duplexes. RNA. 2004;10:1507–1517. doi: 10.1261/rna.5248604.
    1. R Development Core Team. R: A Language and Environment for Statistical Computing. Vienna, Austria. 2009.
    1. Jakymiw A, Pauley KM, Li S, Ikeda K, Lian S, Eystathioy T, Satoh M, Fritzler MJ, Chan EK. The role of GW/P-bodies in RNA processing and silencing. J Cell Sci. 2007;120:1317–1323. doi: 10.1242/jcs.03429.
    1. Komoto J, Usui S, Otsuki S, Terao A. Infantile autism and Duchenne muscular dystrophy. J Autism Dev Disord. 1984;14:191–195. doi: 10.1007/BF02409661.
    1. Zwaigenbaum L, Bryson S, Rogers T, Roberts W, Brian J, Szatmari P. Behavioral manifestations of autism in the first year of life. Int J Dev Neurosci. 2005;23:143–152. doi: 10.1016/j.ijdevneu.2004.05.001.
    1. Horvath K, Papadimitriou JC, Rabsztyn A, Drachenberg C, Tildon JT. Gastrointestinal abnormalities in children with autistic disorder. J Pediatr. 1999;135:559–563. doi: 10.1016/S0022-3476(99)70052-1.
    1. Horvath K, Perman JA. Autism and gastrointestinal symptoms. Curr Gastroenterol Rep. 2002;4:251–258. doi: 10.1007/s11894-002-0071-6.
    1. White JF. Intestinal pathophysiology in autism. Exp Biol Med (Maywood) 2003;228:639–649.
    1. Jyonouchi H, Geng L, Ruby A, Reddy C, Zimmerman-Bier B. Evaluation of an association between gastrointestinal symptoms and cytokine production against common dietary proteins in children with autism spectrum disorders. J Pediatr. 2005;146:605–610. doi: 10.1016/j.jpeds.2005.01.027.
    1. Jyonouchi H, Geng L, Ruby A, Zimmerman-Bier B. Dysregulated innate immune responses in young children with autism spectrum disorders: their relationship to gastrointestinal symptoms and dietary intervention. Neuropsychobiology. 2005;51:77–85. doi: 10.1159/000084164.
    1. Erickson CA, Stigler KA, Corkins MR, Posey DJ, Fitzgerald JF, McDougle CJ. Gastrointestinal factors in autistic disorder: a critical review. J Autism Dev Disord. 2005;35:713–727. doi: 10.1007/s10803-005-0019-4.
    1. Valicenti-McDermott M, McVicar K, Rapin I, Wershil BK, Cohen H, Shinnar S. Frequency of gastrointestinal symptoms in children with autistic spectrum disorders and association with family history of autoimmune disease. J Dev Behav Pediatr. 2006;27(Suppl 2):S128–136. doi: 10.1097/00004703-200604002-00011.
    1. Sempere LF, Freemantle S, Pitha-Rowe I, Moss E, Dmitrovsky E, Ambros V. Expression profiling of mammalian microRNAs uncovers a subset of brain-expressed microRNAs with possible roles in murine and human neuronal differentiation. Genome Biol. 2004;5:R13. doi: 10.1186/gb-2004-5-3-r13.
    1. Schaefer A, O'Carroll D, Tan CL, Hillman D, Sugimori M, Llinas R, Greengard P. Cerebellar neurodegeneration in the absence of microRNAs. J Exp Med. 2007;204:1553–1558. doi: 10.1084/jem.20070823.
    1. Liang Y, Ridzon D, Wong L, Chen C. Characterization of microRNA expression profiles in normal human tissues. BMC Genomics. 2007;8:166. doi: 10.1186/1471-2164-8-166.
    1. Krichevsky AM, King KS, Donahue CP, Khrapko K, Kosik KS. A microRNA array reveals extensive regulation of microRNAs during brain development. RNA. 2003;9:1274–1281. doi: 10.1261/rna.5980303.
    1. John B, Enright AJ, Aravin A, Tuschl T, Sander C, Marks DS. Human MicroRNA targets. PLoS Biol. 2004;2:e363. doi: 10.1371/journal.pbio.0020363.
    1. Friedman RC, Farh KK, Burge CB, Bartel DP. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 2009;19:92–105. doi: 10.1101/gr.082701.108.
    1. Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 2005;120:15–20. doi: 10.1016/j.cell.2004.12.035.
    1. Gennarino VA, Sardiello M, Avellino R, Meola N, Maselli V, Anand S, Cutillo L, Ballabio A, Banfi S. MicroRNA target prediction by expression analysis of host genes. Genome Res. 2009;19:481–490. doi: 10.1101/gr.084129.108.
    1. Saccomani L, Veneselli E, Di Stefano S, Celle ME, De Negri M. Early autism and congenital muscular dystrophy: a clinical case. Pediatr Med Chir. 1992;14:231–233.
    1. Duffield GE, Watson NP, Mantani A, Peirson SN, Robles-Murguia M, Loros JJ, Israel MA, Dunlap JC. A role for Id2 in regulating photic entrainment of the mammalian circadian system. Curr Biol. 2009;19:297–304. doi: 10.1016/j.cub.2008.12.052.
    1. Seeburg DP, Pak D, Sheng M. Polo-like kinases in the nervous system. Oncogene. 2005;24:292–298. doi: 10.1038/sj.onc.1208277.
    1. Seeburg DP, Feliu-Mojer M, Gaiottino J, Pak DT, Sheng M. Critical role of CDK5 and Polo-like kinase 2 in homeostatic synaptic plasticity during elevated activity. Neuron. 2008;58:571–583. doi: 10.1016/j.neuron.2008.03.021.
    1. Seeburg DP, Sheng M. Activity-induced Polo-like kinase 2 is required for homeostatic plasticity of hippocampal neurons during epileptiform activity. J Neurosci. 2008;28:6583–6591. doi: 10.1523/JNEUROSCI.1853-08.2008.
    1. Wimpory D, Nicholas B, Nash S. Social timing, clock genes and autism: a new hypothesis. J Intellect Disabil Res. 2002;46:352–358. doi: 10.1046/j.1365-2788.2002.00423.x.
    1. Nicholas B, Rudrasingham V, Nash S, Kirov G, Owen MJ, Wimpory DC. Association of Per1 and Npas2 with autistic disorder: support for the clock genes/social timing hypothesis. Mol Psychiatry. 2007;12:581–592. doi: 10.1038/sj.mp.4001953.
    1. Zhang J, Fang Z, Jud C, Vansteensel MJ, Kaasik K, Lee CC, Albrecht U, Tamanini F, Meijer JH, Oostra BA, Nelson DL. Fragile X-related proteins regulate mammalian circadian behavioral rhythms. Am J Hum Genet. 2008;83:43–52. doi: 10.1016/j.ajhg.2008.06.003.
    1. Bourgeron T. The possible interplay of synaptic and clock genes in autism spectrum disorders. Cold Spring Harb Symp Quant Biol. 2007;72:645–654. doi: 10.1101/sqb.2007.72.020.
    1. Melke J, Goubran Botros H, Chaste P, Betancur C, Nygren G, Anckarsater H, Rastam M, Stahlberg O, Gillberg IC, Delorme R, Chabane N, Mouren-Simeoni MC, Fauchereau F, Durand CM, Chevalier F, Drouot X, Collet C, Launay JM, Leboyer M, Gillberg C, Bourgeron T. Abnormal melatonin synthesis in autism spectrum disorders. Mol Psychiatry. 2008;13:90–98. doi: 10.1038/sj.mp.4002016.
    1. Hu VW, Sarachana T, Kim KS, Nguyen A, Kulkarni S, Steinberg ME, Luu T, Lai Y, Lee NH. Gene expression profiling differentiates autism case-controls and phenotypic variants of autism spectrum disorders: evidence for circadian rhythm dysfunction in severe autism. Autism Res. 2009;2:78–97. doi: 10.1002/aur.73.
    1. Perry E, Walker M, Grace J, Perry R. Acetylcholine in mind: a neurotransmitter correlate of consciousness? Trends Neurosci. 1999;22:273–280. doi: 10.1016/S0166-2236(98)01361-7.
    1. Perry EK, Lee ML, Martin-Ruiz CM, Court JA, Volsen SG, Merrit J, Folly E, Iversen PE, Bauman ML, Perry RH, Wenk GL. Cholinergic activity in autism: abnormalities in the cerebral cortex and basal forebrain. Am J Psychiatry. 2001;158:1058–1066. doi: 10.1176/appi.ajp.158.7.1058.
    1. Rinaldi T, Kulangara K, Antoniello K, Markram H. Elevated NMDA receptor levels and enhanced postsynaptic long-term potentiation induced by prenatal exposure to valproic acid. Proc Natl Acad Sci USA. 2007;104:13501–13506. doi: 10.1073/pnas.0704391104.
    1. Hussman JP. Suppressed GABAergic inhibition as a common factor in suspected etiologies of autism. J Autism Dev Disord. 2001;31:247–248. doi: 10.1023/A:1010715619091.
    1. Kocerha J, Faghihi MA, Lopez-Toledano MA, Huang J, Ramsey AJ, Caron MG, Sales N, Willoughby D, Elmen J, Hansen HF, Orum H, Kauppinen S, Kenny PJ, Wahlestedt C. MicroRNA-219 modulates NMDA receptor-mediated neurobehavioral dysfunction. Proc Natl Acad Sci USA. 2009;106:3507–3512. doi: 10.1073/pnas.0805854106.
    1. Carlsson ML. Hypothesis: is infantile autism a hypoglutamatergic disorder? Relevance of glutamate - serotonin interactions for pharmacotherapy. J Neural Transm. 1998;105:525–535. doi: 10.1007/s007020050076.
    1. Moy SS, Nadler JJ. Advances in behavioral genetics: mouse models of autism. Mol Psychiatry. 2008;13:4–26. doi: 10.1038/sj.mp.4002082.
    1. Moy SS, Nadler JJ, Poe MD, Nonneman RJ, Young NB, Koller BH, Crawley JN, Duncan GE, Bodfish JW. Development of a mouse test for repetitive, restricted behaviors: relevance to autism. Behav Brain Res. 2008;188:178–194. doi: 10.1016/j.bbr.2007.10.029.
    1. Lehohla M, Kellaway L, Russell VA. NMDA receptor function in the prefrontal cortex of a rat model for attention-deficit hyperactivity disorder. Metab Brain Dis. 2004;19:35–42. doi: 10.1023/B:.
    1. Adams J, Crosbie J, Wigg K, Ickowicz A, Pathare T, Roberts W, Malone M, Schachar R, Tannock R, Kennedy JL, Barr CL. Glutamate receptor, ionotropic, N-methyl D-aspartate 2A (GRIN2A) gene as a positional candidate for attention-deficit/hyperactivity disorder in the 16p13 region. Mol Psychiatry. 2004;9:494–499. doi: 10.1038/sj.mp.4001455.
    1. Beneyto M, Meador-Woodruff JH. Lamina-specific abnormalities of NMDA receptor-associated postsynaptic protein transcripts in the prefrontal cortex in schizophrenia and bipolar disorder. Neuropsychopharmacology. 2008;33:2175–2186. doi: 10.1038/sj.npp.1301604.
    1. Kauselmann G, Weiler M, Wulff P, Jessberger S, Konietzko U, Scafidi J, Staubli U, Bereiter-Hahn J, Strebhardt K, Kuhl D. The polo-like protein kinases Fnk and Snk associate with a Ca(2+)- and integrin-binding protein and are regulated dynamically with synaptic plasticity. EMBO J. 1999;18:5528–5539. doi: 10.1093/emboj/18.20.5528.
    1. Weerdt BC van de, Medema RH. Polo-like kinases: a team in control of the division. Cell Cycle. 2006;5:853–864.
    1. Pak DT, Sheng M. Targeted protein degradation and synapse remodeling by an inducible protein kinase. Science. 2003;302:1368–1373. doi: 10.1126/science.1082475.
    1. Sheng M, Hoogenraad CC. The postsynaptic architecture of excitatory synapses: a more quantitative view. Annu Rev Biochem. 2007;76:823–847. doi: 10.1146/annurev.biochem.76.060805.160029.
    1. Lampe AK, Bushby KM. Collagen VI related muscle disorders. J Med Genet. 2005;42:673–685. doi: 10.1136/jmg.2002.002311.
    1. Lampe AK, Dunn DM, von Niederhausern AC, Hamil C, Aoyagi A, Laval SH, Marie SK, Chu ML, Swoboda K, Muntoni F, Bonnemann CG, Flanigan KM, Bushby KM, Weiss RB. Automated genomic sequence analysis of the three collagen VI genes: applications to Ullrich congenital muscular dystrophy and Bethlem myopathy. J Med Genet. 2005;42:108–120. doi: 10.1136/jmg.2004.023754.
    1. Jobsis GJ, Keizers H, Vreijling JP, de Visser M, Speer MC, Wolterman RA, Baas F, Bolhuis PA. Type VI collagen mutations in Bethlem myopathy, an autosomal dominant myopathy with contractures. Nat Genet. 1996;14:113–115. doi: 10.1038/ng0996-113.
    1. Martoni E, Urciuolo A, Sabatelli P, Fabris M, Bovolenta M, Neri M, Grumati P, D'Amico A, Pane M, Mercuri E, Bertini E, Merlini L, Bonaldo P, Ferlini A, Gualandi F. Identification and characterization of novel collagen VI non-canonical splicing mutations causing Ullrich congenital muscular dystrophy. Hum Mutat. 2009;30:E662–672. doi: 10.1002/humu.21022.
    1. Ishikawa H, Sugie K, Murayama K, Awaya A, Suzuki Y, Noguchi S, Hayashi YK, Nonaka I, Nishino I. Ullrich disease due to deficiency of collagen VI in the sarcolemma. Neurology. 2004;62:620–623.
    1. Baker NL, Morgelin M, Peat R, Goemans N, North KN, Bateman JF, Lamande SR. Dominant collagen VI mutations are a common cause of Ullrich congenital muscular dystrophy. Hum Mol Genet. 2005;14:279–293. doi: 10.1093/hmg/ddi025.
    1. Zwaigenbaum L, Tarnopolsky M. Two children with muscular dystrophies ascertained due to referral for diagnosis of autism. J Autism Dev Disord. 2003;33:193–199. doi: 10.1023/A:1022947728569.
    1. Ming X, Brimacombe M, Wagner GC. Prevalence of motor impairment in autism spectrum disorders. Brain Dev. 2007;29:565–570. doi: 10.1016/j.braindev.2007.03.002.
    1. Heuer JG, Li K, Kaufman TC. The Drosophila homeotic target gene centrosomin (cnn) encodes a novel centrosomal protein with leucine zippers and maps to a genomic region required for midgut morphogenesis. Development. 1995;121:3861–3876.
    1. Verde I, Pahlke G, Salanova M, Zhang G, Wang S, Coletti D, Onuffer J, Jin SL, Conti M. Myomegalin is a novel protein of the golgi/centrosome that interacts with a cyclic nucleotide phosphodiesterase. J Biol Chem. 2001;276:11189–11198. doi: 10.1074/jbc.M006546200.
    1. Kesavapany S, Li BS, Amin N, Zheng YL, Grant P, Pant HC. Neuronal cyclin-dependent kinase 5: role in nervous system function and its specific inhibition by the Cdk5 inhibitory peptide. Biochim Biophys Acta. 2004;1697:143–153.
    1. Mochida GH, Walsh CA. Molecular genetics of human microcephaly. Curr Opin Neurol. 2001;14:151–156. doi: 10.1097/00019052-200104000-00003.
    1. Schumann CM, Barnes CC, Lord C, Courchesne E. Amygdala enlargement in toddlers with autism related to severity of social and communication impairments. Biol Psychiatry. 2009;66:942–949. doi: 10.1016/j.biopsych.2009.07.007.
    1. Redcay E, Courchesne E. When is the brain enlarged in autism? A meta-analysis of all brain size reports. Biol Psychiatry. 2005;58:1–9. doi: 10.1016/j.biopsych.2005.03.026.
    1. Courchesne E, Pierce K. Brain overgrowth in autism during a critical time in development: implications for frontal pyramidal neuron and interneuron development and connectivity. Int J Dev Neurosci. 2005;23:153–170. doi: 10.1016/j.ijdevneu.2005.01.003.
    1. Carper RA, Courchesne E. Localized enlargement of the frontal cortex in early autism. Biol Psychiatry. 2005;57:126–133. doi: 10.1016/j.biopsych.2004.11.005.
    1. Lainhart JE, Piven J, Wzorek M, Landa R, Santangelo SL, Coon H, Folstein SE. Macrocephaly in children and adults with autism. J Am Acad Child Adolesc Psychiatry. 1997;36:282–290. doi: 10.1097/00004583-199702000-00019.
    1. Davis LK, Hazlett HC, Librant AL, Nopoulos P, Sheffield VC, Piven J, Wassink TH. Cortical enlargement in autism is associated with a functional VNTR in the monoamine oxidase A gene. Am J Med Genet B Neuropsychiatr Genet. 2008;147B:1145–1151. doi: 10.1002/ajmg.b.30738.
    1. Hazlett HC, Poe M, Gerig G, Smith RG, Provenzale J, Ross A, Gilmore J, Piven J. Magnetic resonance imaging and head circumference study of brain size in autism: birth through age 2 years. Arch Gen Psychiatry. 2005;62:1366–1376. doi: 10.1001/archpsyc.62.12.1366.
    1. Hazlett HC, Poe MD, Gerig G, Smith RG, Piven J. Cortical gray and white brain tissue volume in adolescents and adults with autism. Biol Psychiatry. 2006;59:1–6. doi: 10.1016/j.biopsych.2005.06.015.
    1. Wassink TH, Hazlett HC, Epping EA, Arndt S, Dager SR, Schellenberg GD, Dawson G, Piven J. Cerebral cortical gray matter overgrowth and functional variation of the serotonin transporter gene in autism. Arch Gen Psychiatry. 2007;64:709–717. doi: 10.1001/archpsyc.64.6.709.
    1. Freitag CM, Luders E, Hulst HE, Narr KL, Thompson PM, Toga AW, Krick C, Konrad C. Total brain volume and corpus callosum size in medication-naive adolescents and young adults with autism spectrum disorder. Biol Psychiatry. 2009;66:316–319. doi: 10.1016/j.biopsych.2009.03.011.
    1. Fombonne E, Roge B, Claverie J, Courty S, Fremolle J. Microcephaly and macrocephaly in autism. J Autism Dev Disord. 1999;29:113–119. doi: 10.1023/A:1023036509476.
    1. Piven J, Arndt S, Bailey J, Havercamp S, Andreasen NC, Palmer P. An MRI study of brain size in autism. Am J Psychiatry. 1995;152:1145–1149.
    1. Piven J, Arndt S, Bailey J, Andreasen N. Regional brain enlargement in autism: a magnetic resonance imaging study. J Am Acad Child Adolesc Psychiatry. 1996;35:530–536.
    1. Woodhouse W, Bailey A, Rutter M, Bolton P, Baird G, Le Couteur A. Head circumference in autism and other pervasive developmental disorders. J Child Psychol Psychiatry. 1996;37:665–671. doi: 10.1111/j.1469-7610.1996.tb01458.x.
    1. Kemper TL, Bauman M. Neuropathology of infantile autism. J Neuropathol Exp Neurol. 1998;57:645–652. doi: 10.1097/00005072-199807000-00001.
    1. Saba R, Goodman CD, Huzarewich RL, Robertson C, Booth SA. A miRNA signature of prion induced neurodegeneration. PLoS One. 2008;3:e3652. doi: 10.1371/journal.pone.0003652.
    1. Wilfred BR, Wang WX, Nelson PT. Energizing miRNA research: a review of the role of miRNAs in lipid metabolism, with a prediction that miR-103/107 regulates human metabolic pathways. Mol Genet Metab. 2007;91:209–217. doi: 10.1016/j.ymgme.2007.03.011.
    1. Kotzbauer PT, Truax AC, Trojanowski JQ, Lee VM. Altered neuronal mitochondrial coenzyme A synthesis in neurodegeneration with brain iron accumulation caused by abnormal processing, stability, and catalytic activity of mutant pantothenate kinase 2. J Neurosci. 2005;25:689–698. doi: 10.1523/JNEUROSCI.4265-04.2005.
    1. Zhang YM, Rock CO, Jackowski S. Biochemical properties of human pantothenate kinase 2 isoforms and mutations linked to pantothenate kinase-associated neurodegeneration. J Biol Chem. 2006;281:107–114. doi: 10.1074/jbc.M508825200.
    1. Vancassel S, Durand G, Barthelemy C, Lejeune B, Martineau J, Guilloteau D, Andres C, Chalon S. Plasma fatty acid levels in autistic children. Prostaglandins Leukot Essent Fatty Acids. 2001;65:1–7. doi: 10.1054/plef.2001.0281.
    1. Johnson KP, Malow BA. Sleep in children with autism spectrum disorders. Curr Treat Options Neurol. 2008;10:350–359. doi: 10.1007/s11940-008-0038-5.
    1. Malow BA. Sleep disorders, epilepsy, and autism. Ment Retard Dev Disabil Res Rev. 2004;10:122–125. doi: 10.1002/mrdd.20023.
    1. Hu VW, Steinberg ME. Novel clustering of items from the Autism Diagnostic Interview-Revised to define phenotypes within autism spectrum disorders. Autism Res. 2009;2:67–77. doi: 10.1002/aur.72.
    1. Moore RY, Eichler VB. Loss of a circadian adrenal corticosterone rhythm following suprachiasmatic lesions in the rat. Brain Res. 1972;42:201–206. doi: 10.1016/0006-8993(72)90054-6.
    1. Ralph MR, Foster RG, Davis FC, Menaker M. Transplanted suprachiasmatic nucleus determines circadian period. Science. 1990;247:975–978. doi: 10.1126/science.2305266.
    1. Cheng HY, Obrietan K. Revealing a role of microRNAs in the regulation of the biological clock. Cell Cycle. 2007;6:3034–3035.
    1. Cheng HY, Papp JW, Varlamova O, Dziema H, Russell B, Curfman JP, Nakazawa T, Shimizu K, Okamura H, Impey S, Obrietan K. microRNA modulation of circadian-clock period and entrainment. Neuron. 2007;54:813–829. doi: 10.1016/j.neuron.2007.05.017.
    1. Peddada S, Yasui DH, LaSalle JM. Inhibitors of differentiation (ID1, ID2, ID3 and ID4) genes are neuronal targets of MeCP2 that are elevated in Rett syndrome. Hum Mol Genet. 2006;15:2003–2014. doi: 10.1093/hmg/ddl124.
    1. Herbert MR, Russo JP, Yang S, Roohi J, Blaxill M, Kahler SG, Cremer L, Hatchwell E. Autism and environmental genomics. Neurotoxicology. 2006;27:671–684. doi: 10.1016/j.neuro.2006.03.017.
    1. Levy SE, Souders MC, Ittenbach RF, Giarelli E, Mulberg AE, Pinto-Martin JA. Relationship of dietary intake to gastrointestinal symptoms in children with autistic spectrum disorders. Biol Psychiatry. 2007;61:492–497. doi: 10.1016/j.biopsych.2006.07.013.
    1. Ingudomnukul E, Baron-Cohen S, Wheelwright S, Knickmeyer R. Elevated rates of testosterone-related disorders in women with autism spectrum conditions. Horm Behav. 2007;51:597–604. doi: 10.1016/j.yhbeh.2007.02.001.
    1. Knickmeyer RC, Baron-Cohen S. Fetal testosterone and sex differences in typical social development and in autism. J Child Neurol. 2006;21:825–845. doi: 10.1177/08830738060210101601.
    1. Zamani MR, Desmond NL, Levy WB. Estradiol modulates long-term synaptic depression in female rat hippocampus. J Neurophysiol. 2000;84:1800–1808.
    1. MacLusky NJ, Hajszan T, Prange-Kiel J, Leranth C. Androgen modulation of hippocampal synaptic plasticity. Neuroscience. 2006;138:957–965. doi: 10.1016/j.neuroscience.2005.12.054.
    1. Harris SW, Hessl D, Goodlin-Jones B, Ferranti J, Bacalman S, Barbato I, Tassone F, Hagerman PJ, Herman H, Hagerman RJ. Autism profiles of males with fragile X syndrome. Am J Ment Retard. 2008;113:427–438. doi: 10.1352/2008.113:427-438.
    1. Talebizadeh Z, Lam DY, Theodoro MF, Bittel DC, Lushington GH, Butler MG. Novel splice isoforms for NLGN3 and NLGN4 with possible implications in autism. J Med Genet. 2006;43:e21. doi: 10.1136/jmg.2005.036897.
    1. Baron CA, Liu SY, Hicks C, Gregg JP. Utilization of lymphoblastoid cell lines as a system for the molecular modeling of autism. J Autism Dev Disord. 2006;36:973–982. doi: 10.1007/s10803-006-0134-x.
    1. Schule B, Li HH, Fisch-Kohl C, Purmann C, Francke U. DLX5 and DLX6 expression is biallelic and not modulated by MeCP2 deficiency. Am J Hum Genet. 2007;81:492–506. doi: 10.1086/520063.
    1. Brennecke J, Stark A, Russell RB, Cohen SM. Principles of microRNA-target recognition. PLoS Biol. 2005;3:e85. doi: 10.1371/journal.pbio.0030085.
    1. Krek A, Grun D, Poy MN, Wolf R, Rosenberg L, Epstein EJ, MacMenamin P, da Piedade I, Gunsalus KC, Stoffel M, Rajewsky N. Combinatorial microRNA target predictions. Nat Genet. 2005;37:495–500. doi: 10.1038/ng1536.
    1. Lai EC. Predicting and validating microRNA targets. Genome Biol. 2004;5:115. doi: 10.1186/gb-2004-5-9-115.
    1. Lewis BP, Shih IH, Jones-Rhoades MW, Bartel DP, Burge CB. Prediction of mammalian microRNA targets. Cell. 2003;115:787–798. doi: 10.1016/S0092-8674(03)01018-3.

Source: PubMed

3
Subskrybuj