101
|
Wang X, Sun Q, McGrath SD, Mardis ER, Soloway PD, Clark AG. Transcriptome-wide identification of novel imprinted genes in neonatal mouse brain. PLoS One 2008; 3:e3839. [PMID: 19052635 PMCID: PMC2585789 DOI: 10.1371/journal.pone.0003839] [Citation(s) in RCA: 148] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2008] [Accepted: 11/05/2008] [Indexed: 11/19/2022] Open
Abstract
Imprinted genes display differential allelic expression in a manner that depends on the sex of the transmitting parent. The degree of imprinting is often tissue-specific and/or developmental stage-specific, and may be altered in some diseases including cancer. Here we applied Illumina/Solexa sequencing of the transcriptomes of reciprocal F1 mouse neonatal brains and identified 26 genes with parent-of-origin dependent differential allelic expression. Allele-specific Pyrosequencing verified 17 of them, including three novel imprinted genes. The known and novel imprinted genes all are found in proximity to previously reported differentially methylated regions (DMRs). Ten genes known to be imprinted in placenta had sufficient expression levels to attain a read depth that provided statistical power to detect imprinting, and yet all were consistent with non-imprinting in our transcript count data for neonatal brain. Three closely linked and reciprocally imprinted gene pairs were also discovered, and their pattern of expression suggests transcriptional interference. Despite the coverage of more than 5000 genes, this scan only identified three novel imprinted refseq genes in neonatal brain, suggesting that this tissue is nearly exhaustively characterized. This approach has the potential to yield an complete catalog of imprinted genes after application to multiple tissues and developmental stages, shedding light on the mechanism, bioinformatic prediction, and evolution of imprinted genes and diseases associated with genomic imprinting.
Collapse
Affiliation(s)
- Xu Wang
- Department of Molecular Biology & Genetics, Cornell University, Ithaca, New York, United States of America
| | - Qi Sun
- Computational Biology Service Unit, Life Sciences Core Laboratories Center, Cornell University, Ithaca, New York, United States of America
| | - Sean D. McGrath
- The Genome Center at Washington University, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Elaine R. Mardis
- The Genome Center at Washington University, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Paul D. Soloway
- Division of Nutritional Sciences, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, United States of America
| | - Andrew G. Clark
- Department of Molecular Biology & Genetics, Cornell University, Ithaca, New York, United States of America
- * E-mail:
| |
Collapse
|
102
|
Desforges M, Mynett KJ, Jones RL, Greenwood SL, Westwood M, Sibley CP, Glazier JD. The SNAT4 isoform of the system A amino acid transporter is functional in human placental microvillous plasma membrane. J Physiol 2008; 587:61-72. [PMID: 19015196 PMCID: PMC2667314 DOI: 10.1113/jphysiol.2008.161331] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Placental system A activity is important for the supply of neutral amino acids needed for fetal growth. There are three system A isoforms: SNAT1, SNAT2 and SNAT4, but the contribution of each to system A-mediated transport is unknown. Here, we have used immunohistochemistry to demonstrate that all three isoforms are present in the syncytiotrophoblast suggesting each plays a role in amino acid transport across the placenta. We next tested the hypothesis that the SNAT4 isoform is functional in microvillous plasma membrane vesicles (MVM) from normal human placenta using a method which exploits the unique property of SNAT4 to transport both cationic amino acids as well as the system A-specific substrate MeAIB. The data show that SNAT4 contribution to system A-specific amino acid transport across MVM is higher in first trimester placenta compared to term (approx. 70% and 33%, respectively, P < 0.01). Further experiments performed under more physiological conditions using intact placental villous fragments suggest a contribution of SNAT4 to system A activity in first trimester placenta but minimal contribution at term. In agreement, Western blotting revealed that SNAT4 protein expression is higher in first trimester MVM compared to term (P < 0.05). This study provides the first evidence of SNAT4 activity in human placenta and demonstrates the contribution of SNAT4 to system A-mediated transport decreases between first trimester and term: our data lead us to speculate that at later stages of gestation SNAT1 and/or SNAT2 are more important for the supply of amino acids required for normal fetal growth.
Collapse
Affiliation(s)
- M Desforges
- Maternal and Fetal Health Research Group, University of Manchester, St Mary's Hospital, Hathersage Road, Manchester M13 0JH, UK.
| | | | | | | | | | | | | |
Collapse
|
103
|
Mo B, Callegari E, Telefont M, Renner KJ. Estrogen regulation of proteins in the rat ventromedial nucleus of the hypothalamus. J Proteome Res 2008; 7:5040-8. [PMID: 18841879 DOI: 10.1021/pr8005974] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The effects of estradiol (E2) on the expression of proteins in the pars lateralis of the ventromedial nucleus of the hypothalamus (VMNpl) in ovariectomized rats was studied using 2-dimensional gel electrophoresis followed by RPLC-nanoESI-MS/MS. E2 treatment resulted in the up-regulation of 29 identified proteins. Many of these proteins are implicated in the promotion of neuronal plasticity and signaling.
Collapse
Affiliation(s)
- Bing Mo
- Department of Biology and Neuroscience Group, University of South Dakota, Vermillion, South Daklota 57069, USA
| | | | | | | |
Collapse
|
104
|
Allan MF, Kuehn LA, Cushman RA, Snelling WM, Echternkamp SE, Thallman RM. Confirmation of quantitative trait loci using a low-density single nucleotide polymorphism map for twinning and ovulation rate on bovine chromosome 5. J Anim Sci 2008; 87:46-56. [PMID: 18791147 DOI: 10.2527/jas.2008-0959] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Traditional genetic selection in cattle for traits with low heritability, such as reproduction, has had very little success. With the addition of DNA technologies to the genetic selection toolbox for livestock, the opportunity may exist to improve reproductive efficiency more rapidly in cattle. The US Meat Animal Research Center Production Efficiency Population has 9,186 twinning and 29,571 ovulation rate records for multiple generations of animals, but a significant number of these animals do not have tissue samples available for DNA genotyping. The objectives of this study were to confirm QTL for twinning and ovulation rate previously found on BTA5 and to evaluate the ability of GenoProb to predict genotypic information in a pedigree containing 16,035 animals when using genotypes for 24 SNP from 3 data sets containing 48, 724, or 2,900 animals. Marker data for 21 microsatellites on BTA5 with 297 to 3,395 animals per marker were used in conjunction with each data set of genotyped animals. Genotypic probabilities for females were used to calculate independent variables for regressions of additive, dominance, and imprinting effects. Genotypic regressions were fitted as fixed effects in a 2-trait mixed model analysis by using multiple-trait derivative-free REML. Each SNP was analyzed individually, followed by backward selection fitting all individually significant SNP simultaneously and then removing the least significant SNP until only significant SNP were left. Five significant SNP associations were detected for twinning rate and 3 were detected for ovulation rate. Two of these SNP, 1 for each trait, were significant for imprinting. Additional modeling of paternal and maternal allelic effects confirmed the initial results of imprinting done by contrasting heterozygotes. These results are supported by comparative mapping of mouse and human imprinted genes to this region of bovine chromosome 5.
Collapse
Affiliation(s)
- M F Allan
- USDA, ARS, US Meat Animal Research Center, Clay Center, NE 68933, USA
| | | | | | | | | | | |
Collapse
|
105
|
Maternally inherited Birk Barel mental retardation dysmorphism syndrome caused by a mutation in the genomically imprinted potassium channel KCNK9. Am J Hum Genet 2008; 83:193-9. [PMID: 18678320 DOI: 10.1016/j.ajhg.2008.07.010] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2008] [Revised: 06/18/2008] [Accepted: 07/02/2008] [Indexed: 12/24/2022] Open
Abstract
We describe a maternally transmitted genomic-imprinting syndrome of mental retardation, hypotonia, and unique dysmorphism with elongated face. We mapped the disease-associated locus to approximately 7.27 Mb on chromosome 8q24 and demonstrated that the disease is caused by a missense mutation in the maternal copy of KCNK9 within this locus. KCNK9 is maternally transmitted (imprinted with paternal silencing) and encodes K(2P)9.1, a member of the two pore-domain potassium channel (K(2P)) subfamily. The mutation fully abolishes the channel's currents--both when functioning as a homodimer or as a heterodimer with K(2P)3.1.
Collapse
|
106
|
Jamieson SE, de Roubaix LA, Cortina-Borja M, Tan HK, Mui EJ, Cordell HJ, Kirisits MJ, Miller EN, Peacock CS, Hargrave AC, Coyne JJ, Boyer K, Bessieres MH, Buffolano W, Ferret N, Franck J, Kieffer F, Meier P, Nowakowska DE, Paul M, Peyron F, Stray-Pedersen B, Prusa AR, Thulliez P, Wallon M, Petersen E, McLeod R, Gilbert RE, Blackwell JM. Genetic and epigenetic factors at COL2A1 and ABCA4 influence clinical outcome in congenital toxoplasmosis. PLoS One 2008; 3:e2285. [PMID: 18523590 PMCID: PMC2390765 DOI: 10.1371/journal.pone.0002285] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2008] [Accepted: 04/11/2008] [Indexed: 01/26/2023] Open
Abstract
Background Primary Toxoplasma gondii infection during pregnancy can be transmitted to the fetus. At birth, infected infants may have intracranial calcification, hydrocephalus, and retinochoroiditis, and new ocular lesions can occur at any age after birth. Not all children who acquire infection in utero develop these clinical signs of disease. Whilst severity of disease is influenced by trimester in which infection is acquired by the mother, other factors including genetic predisposition may contribute. Methods and Findings In 457 mother-child pairs from Europe, and 149 child/parent trios from North America, we show that ocular and brain disease in congenital toxoplasmosis associate with polymorphisms in ABCA4 encoding ATP-binding cassette transporter, subfamily A, member 4. Polymorphisms at COL2A1 encoding type II collagen associate only with ocular disease. Both loci showed unusual inheritance patterns for the disease allele when comparing outcomes in heterozygous affected children with outcomes in affected children of heterozygous mothers. Modeling suggested either an effect of mother's genotype, or parent-of-origin effects. Experimental studies showed that both ABCA4 and COL2A1 show isoform-specific epigenetic modifications consistent with imprinting. Conclusions These associations between clinical outcomes of congenital toxoplasmosis and polymorphisms at ABCA4 and COL2A1 provide novel insight into the molecular pathways that can be affected by congenital infection with this parasite.
Collapse
Affiliation(s)
- Sarra E. Jamieson
- Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Lee-Anne de Roubaix
- Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Mario Cortina-Borja
- Centre for Paediatric Epidemiology and Biostatistics, Institute of Child Health, University College London, London, United Kingdom
| | - Hooi Kuan Tan
- Centre for Paediatric Epidemiology and Biostatistics, Institute of Child Health, University College London, London, United Kingdom
| | - Ernest J. Mui
- Departments of Ophthalmology, Medicine, Pediatrics, Committees on Immunology, Molecular Medicine, and Genetics, University of Chicago, and Michael Reese Hospital and Medical Center, Chicago, Illinois, United States of America
| | - Heather J. Cordell
- Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrookes Hospital, Cambridge, United Kingdom
- Institute of Human Genetics, Newcastle University, International Centre for Life, Newcastle upon Tyne, United Kingdom
| | - Michael J. Kirisits
- Departments of Ophthalmology, Medicine, Pediatrics, Committees on Immunology, Molecular Medicine, and Genetics, University of Chicago, and Michael Reese Hospital and Medical Center, Chicago, Illinois, United States of America
| | - E. Nancy Miller
- Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Christopher S. Peacock
- Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Aubrey C. Hargrave
- Departments of Ophthalmology, Medicine, Pediatrics, Committees on Immunology, Molecular Medicine, and Genetics, University of Chicago, and Michael Reese Hospital and Medical Center, Chicago, Illinois, United States of America
| | - Jessica J. Coyne
- Departments of Ophthalmology, Medicine, Pediatrics, Committees on Immunology, Molecular Medicine, and Genetics, University of Chicago, and Michael Reese Hospital and Medical Center, Chicago, Illinois, United States of America
| | - Kenneth Boyer
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Rush University Medical Center, Chicago, Illinois, United States of America
| | | | - Wilma Buffolano
- Department of Paediatrics, University of Naples "Frederico II", Naples, Italy
| | - Nicole Ferret
- Service de Parasitologie et Mycologie, Hopital Archet II, Nice, France
| | | | - François Kieffer
- Department of Paediatrics, Institut de Puériculture, Paris, France
| | - Paul Meier
- Department of Biostatistics, Columbia University, New York, New York, United States of America
| | - Dorota E. Nowakowska
- Department of Fetal-Maternal Medicine and Gynecology, Medical University, Lodz, Rzgowska, Poland
| | - Malgorzata Paul
- Department and Clinic of Tropical and Parasitic Diseases, University of Medical Sciences, Poznań, Poland
| | - François Peyron
- Hospices Civils de Lyon, Service de Parasitologie, Hôpital de la Croix-Rousse, Lyon, France
| | - Babill Stray-Pedersen
- Department of Obstetrics and Gynaecology, University of Oslo, Rikshospitalet-Radiumhospitalet, Sognsvannsvn, Oslo, Norway
| | - Andrea-Romana Prusa
- Department of Pediatrics, Division of Neonatology, Congenital Disorders and Intensive Care, Medical University of Vienna, Vienna, Austria
| | | | - Martine Wallon
- Hospices Civils de Lyon, Service de Parasitologie, Hôpital de la Croix-Rousse, Lyon, France
| | - Eskild Petersen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Rima McLeod
- Departments of Ophthalmology, Medicine, Pediatrics, Committees on Immunology, Molecular Medicine, and Genetics, University of Chicago, and Michael Reese Hospital and Medical Center, Chicago, Illinois, United States of America
| | - Ruth E. Gilbert
- Centre for Paediatric Epidemiology and Biostatistics, Institute of Child Health, University College London, London, United Kingdom
| | - Jenefer M. Blackwell
- Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrookes Hospital, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
107
|
Cheverud JM, Hager R, Roseman C, Fawcett G, Wang B, Wolf JB. Genomic imprinting effects on adult body composition in mice. Proc Natl Acad Sci U S A 2008; 105:4253-8. [PMID: 18337500 PMCID: PMC2393747 DOI: 10.1073/pnas.0706562105] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2007] [Indexed: 11/18/2022] Open
Abstract
Genomic imprinting results in the differential expression of genes, depending on which allele is inherited from the mother and which from the father. The effects of such differential gene expression are reflected in phenotypic differences between the reciprocal heterozygotes (Aa vs. aA). Although many imprinted genes have been identified and play a key role in development, little is known about the contribution of imprinting to quantitative variation in trait expression. Here, we examine this problem by mapping imprinting effects on adult body composition traits in the F(3) generation of an intercross between the Large (LG/J) and Small (SM/J) inbred mouse strains. We identified eight pleiotropic imprinted quantitative trait loci (iQTL) located throughout the genome. Most iQTL are in novel locations that have not previously been associated with imprinting effects, but those on chromosomes 7, 12, and centromeric 18 lie in regions previously identified as containing imprinted genes. Our results show that the effects of genomic imprinting are relatively small, with reciprocal heterozygotes differing by approximately 0.25 standard deviation units and the effects at each locus accounting for 1% to 4% of the phenotypic variance. We detected a variety of imprinting patterns, with paternal expression being the most common. These results indicate that genomic imprinting has small, but detectable, effects on the normal variation of complex traits in adults and is likely to be more common than usually thought.
Collapse
Affiliation(s)
- James M. Cheverud
- *Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Reinmar Hager
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom; and
| | - Charles Roseman
- Department of Anthropology, University of Illinois at Urbana–Champaign, Urbana, IL 61801
| | - Gloria Fawcett
- *Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Bing Wang
- *Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Jason B. Wolf
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom; and
| |
Collapse
|
108
|
Methylation perturbations in retroelements within the genome of a Mus interspecific hybrid correlate with double minute chromosome formation. Genomics 2008; 91:267-73. [DOI: 10.1016/j.ygeno.2007.12.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2007] [Revised: 10/10/2007] [Accepted: 12/05/2007] [Indexed: 12/22/2022]
|
109
|
Ruf N, Bähring S, Galetzka D, Pliushch G, Luft FC, Nürnberg P, Haaf T, Kelsey G, Zechner U. Sequence-based bioinformatic prediction and QUASEP identify genomic imprinting of the KCNK9 potassium channel gene in mouse and human. Hum Mol Genet 2007; 16:2591-9. [PMID: 17704508 DOI: 10.1093/hmg/ddm216] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Genomic imprinting is the epigenetic marking of gene subsets resulting in monoallelic or predominant expression of one of the two parental alleles according to their parental origin. We describe the systematic experimental verification of a prioritized 16 candidate imprinted gene set predicted by sequence-based bioinformatic analyses. We used Quantification of Allele-Specific Expression by Pyrosequencing (QUASEP) and discovered maternal-specific imprinted expression of the Kcnk9 gene as well as strain-dependent preferential expression of the Rarres1 gene in E11.5 (C57BL/6 x Cast/Ei)F1 and informative (C57BL/6 x Cast/Ei) x C57BL/6 backcross mouse embryos. For the remaining 14 candidate imprinted genes, we observed biallelic expression. In adult mouse tissues, we found that Kcnk9 expression was restricted to the brain and also was maternal-specific. QUASEP analysis of informative human fetal brain samples further demonstrated maternal-specific imprinted expression of the human KCNK9 orthologue. The CpG islands associated with the mouse and human Kcnk9/KCNK9 genes were not differentially methylated, but strongly hypomethylated. Thus, we speculate that mouse Kcnk9 imprinting may be regulated by the maternal germline differentially methylated region in Peg13, an imprinted non-coding RNA gene in close proximity to Kcnk9 on distal mouse chromosome 15. Our data have major implications for the proposed role of Kcnk9 in neurodevelopment, apoptosis and tumourigenesis, as well as for the efficiency of sequence-based bioinformatic predictions of novel imprinted genes.
Collapse
Affiliation(s)
- Nico Ruf
- Max-Delbrueck-Center for Molecular Medicine, D-13125 Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
110
|
Attia M, Rachez C, De Pauw A, Avner P, Rogner UC. Nap1l2 promotes histone acetylation activity during neuronal differentiation. Mol Cell Biol 2007; 27:6093-102. [PMID: 17591696 PMCID: PMC1952155 DOI: 10.1128/mcb.00789-07] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The deletion of the neuronal Nap1l2 (nucleosome assembly protein 1-like 2) gene in mice causes neural tube defects. We demonstrate here that this phenotype correlates with deficiencies in differentiation and increased maintenance of the neural stem cell stage. Nap1l2 associates with chromatin and interacts with histones H3 and H4. Loss of Nap1l2 results in decreased histone acetylation activity, leading to transcriptional changes in differentiating neurons, which include the marked downregulation of the Cdkn1c (cyclin-dependent kinase inhibitor 1c) gene. Cdkn1c expression normally increases during neuronal differentiation, and this correlates with the specific recruitment of the Nap1l2 protein and an increase in acetylated histone H3K9/14 at the site of Cdkn1c transcription. These results lead us to suggest that the Nap1l2 protein plays an important role in regulating transcription in developing neurons via the control of histone acetylation. Our data support the idea that neuronal nucleosome assembly proteins mediate cell-type-specific mechanisms of establishment/modification of a chromatin-permissive state that can affect neurogenesis and neuronal survival.
Collapse
Affiliation(s)
- Mikaël Attia
- Unité Génétique Moléculaire Murine, CNRS URA 2578, Institut Pasteur, 25 rue du Docteur Roux, 75724 Paris Cedex 15, France
| | | | | | | | | |
Collapse
|
111
|
Edwards CA, Ferguson-Smith AC. Mechanisms regulating imprinted genes in clusters. Curr Opin Cell Biol 2007; 19:281-9. [PMID: 17467259 DOI: 10.1016/j.ceb.2007.04.013] [Citation(s) in RCA: 303] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2007] [Accepted: 04/16/2007] [Indexed: 12/20/2022]
Abstract
Clustered imprinted genes are regulated by differentially methylated imprinting control regions (ICRs) that affect gene activity and repression in cis over a large region. Although a primary imprint signal for each of these clusters is DNA methylation, different mechanisms are used to establish and maintain these marks. The majority of ICRs are methylated in the maternal germline and are usually promoters for antisense transcripts whose elongation is associated with imprinting control in the domain. In contrast, ICRs methylated in the paternal germline do not appear to act as promoters and are located between genes. At least one, at the Igf2/H19 locus, is known to function as an insulator. Analysis of ICRs suggests that maternal and paternal methylation imprints function in distinct ways.
Collapse
Affiliation(s)
- Carol A Edwards
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | | |
Collapse
|
112
|
Abstract
Homologues of nucleosome assembly protein 1 (NAP1) have been identified in all eukaryotes. Although initially identified as histone chaperones and chromatin-assembly factors, additional functions include roles in tissue-specific transcription regulation, apoptosis, histone shuttling, and cell-cycle regulation, and extend beyond those of a simple chaperone and assembly factor. NAP1 family members share a structurally conserved fold, the NAP domain. Here we review current knowledge of the NAP family of proteins within the context of the recently determined crystal structure of the NAP1 family's first representative, NAP1 from yeast.
Collapse
Affiliation(s)
- Young-Jun Park
- Howard Hughes Medical Institute and Department of Biochemistry and Molecular Biology, CO State University, Fort Collins, CO 80523-1870, USA.
| | | |
Collapse
|
113
|
Abstract
Imprinted genes are monoallelically expressed in a parent-of-origin-specific manner, but for many genes reported to be imprinted, the occurrence of preferential expression--where both alleles are expressed but one is expressed more strongly than the other in a parent-of-origin-specific way--has been reported. This preferential expression found in genes described as imprinted has not been thoroughly addressed in genomic imprinting studies. To study this phenomenon, 50 genes, reported to be imprinted in the mouse, were chosen for investigation. Preferential expression was observed for 21 of 27 maternally expressed genes. However, only 5 of 23 paternally expressed genes showed preferential expression. Recently, it has been reported that a remarkable proportion of non-imprinted genes show differential allelic expression. If there is overlap between non-imprinted genes that are differentially expressed and imprinted genes that are preferentially expressed, we need to set new definitions of imprinted genes that, in turn, would probably lead to reassessments of the total number of imprinted genes in mammalian species.
Collapse
Affiliation(s)
- Hasan Khatib
- Department of Dairy Science, 1675 Observatory Drive, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|
114
|
Abstract
The epigenetic events that occur during the development of the mammalian embryo are essential for correct gene expression and cell-lineage determination. Imprinted genes are expressed from only one parental allele due to differential epigenetic marks that are established during gametogenesis. Several theories have been proposed to explain the role that genomic imprinting has played over the course of mammalian evolution, but at present it is not clear if a single hypothesis can fully account for the diversity of roles that imprinted genes play. In this review, we discuss efforts to define the extent of imprinting in the mouse genome, and suggest that different imprinted loci may have been wrought by distinct evolutionary forces. We focus on a group of small imprinted domains, which consist of paternally expressed genes embedded within introns of multiexonic transcripts, to discuss the evolution of imprinting at these loci.
Collapse
|
115
|
Wood AJ, Roberts RG, Monk D, Moore GE, Schulz R, Oakey RJ. A screen for retrotransposed imprinted genes reveals an association between X chromosome homology and maternal germ-line methylation. PLoS Genet 2006; 3:e20. [PMID: 17291163 PMCID: PMC1796624 DOI: 10.1371/journal.pgen.0030020] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2006] [Accepted: 12/18/2006] [Indexed: 11/24/2022] Open
Abstract
Imprinted genes undergo epigenetic modifications during gametogenesis, which lead to transcriptional silencing of either the maternally or the paternally derived allele in the subsequent generation. Previous work has suggested an association between imprinting and the products of retrotransposition, but the nature of this link is not well defined. In the mouse, three imprinted genes have been described that originated by retrotransposition and overlap CpG islands which undergo methylation during oogenesis. Nap1l5, U2af1-rs1, and Inpp5f_v2 are likely to encode proteins and share two additional genetic properties: they are located within introns of host transcripts and are derived from parental genes on the X chromosome. Using these sequence features alone, we identified Mcts2, a novel candidate imprinted retrogene on mouse Chromosome 2. Mcts2 has been validated as imprinted by demonstrating that it is paternally expressed and undergoes promoter methylation during oogenesis. The orthologous human retrogenes NAP1L5, INPP5F_V2, and MCTS2 are also shown to be paternally expressed, thus delineating novel imprinted loci on human Chromosomes 4, 10, and 20. The striking correlation between imprinting and X chromosome provenance suggests that retrotransposed elements with homology to the X chromosome can be selectively targeted for methylation during mammalian oogenesis. The conventional view is that DNA carries all of our heritable information and our genes control development into adulthood. The discovery of epigenetics, a term coined to describe effects that are not coded for by DNA sequence, but can nonetheless affect our development and well-being, has added another layer of complexity to our understanding of genetics. One class of genes under epigenetic control are imprinted genes. Mammals inherit two copies of every gene, one from mother and one from father, and in most cases, both are active. However, for a small number of imprinted genes in mammals, only one is active, either the maternal or the paternal copy. Epigenetics amounts to a control system for switching genes on and off appropriately. We focus on a group of little-studied imprinted genes that share features that give clues to their evolutionary origins. These so-called “retrogenes” are protein-coding sequences of DNA that have undergone duplication and jumped into novel locations in the genome. Because of this, it is possible to determine where, and roughly when, many of the imprinted retrogenes originated. This provides an opportunity to study the molecular events that have generated imprinted genes during mammalian evolution.
Collapse
Affiliation(s)
- Andrew J Wood
- Department of Medical and Molecular Genetics, King's College London, London, United Kingdom
| | - Roland G Roberts
- Department of Medical and Molecular Genetics, King's College London, London, United Kingdom
| | - David Monk
- Unit of Clinical and Molecular Genetics, Institute of Child Health, London, United Kingdom
| | - Gudrun E Moore
- Unit of Clinical and Molecular Genetics, Institute of Child Health, London, United Kingdom
| | - Reiner Schulz
- Department of Medical and Molecular Genetics, King's College London, London, United Kingdom
| | - Rebecca J Oakey
- Department of Medical and Molecular Genetics, King's College London, London, United Kingdom
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
116
|
Abstract
The autism spectrum disorders (ASD) comprise a complex group of behaviorally related disorders that are primarily genetic in origin. Involvement of epigenetic regulatory mechanisms in the pathogenesis of ASD has been suggested by the occurrence of ASD in patients with disorders arising from epigenetic mutations (fragile X syndrome) or that involve key epigenetic regulatory factors (Rett syndrome). Moreover, the most common recurrent cytogenetic abnormalities in ASD involve maternally derived duplications of the imprinted domain on chromosome 15q11-13. Thus, parent of origin effects on sharing and linkage to imprinted regions on chromosomes 15q and 7q suggest that these regions warrant specific examination from an epigenetic perspective, particularly because epigenetic modifications do not change the primary genomic sequence, allowing risk epialleles to evade detection using standard screening strategies. This review examines the potential role of epigenetic factors in the etiology of ASD.
Collapse
Affiliation(s)
- N Carolyn Schanen
- Center for Pediatric Research, Nemours Biomedical Research, Wilmington, DE 19803, USA.
| |
Collapse
|
117
|
Zaitoun I, Khatib H. Assessment of genomic imprinting of SLC38A4, NNAT, NAP1L5, and H19 in cattle. BMC Genet 2006; 7:49. [PMID: 17064418 PMCID: PMC1629023 DOI: 10.1186/1471-2156-7-49] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2006] [Accepted: 10/25/2006] [Indexed: 01/09/2023] Open
Abstract
Background At present, few imprinted genes have been reported in cattle compared to human and mouse. Comparative expression analysis and imprinting status are powerful tools for investigating the biological significance of genomic imprinting and studying the regulation mechanisms of imprinted genes. The objective of this study was to assess the imprinting status and pattern of expression of the SLC38A4, NNAT, NAP1L5, and H19 genes in bovine tissues. Results A polymorphism-based approach was used to assess the imprinting status of four bovine genes in a total of 75 tissue types obtained from 12 fetuses and their dams. In contrast to mouse Slc38a4, which is imprinted in a tissue-specific manner, we found that SLC38A4 is not imprinted in cattle, and we found it expressed in all adult tissues examined. Two single nucleotide polymorphisms (SNPs) were identified in NNAT and used to distinguish between monoallelic and biallelic expression in fetal and adult tissues. The two transcripts of NNAT showed paternal expression like their orthologues in human and mouse. However, in contrast to human and mouse, NNAT was expressed in a wide range of tissues, both fetal and adult. Expression analysis of NAP1L5 in five heterozygous fetuses showed that the gene was paternally expressed in all examined tissues, in contrast to mouse where imprinting is tissue-specific. H19 was found to be maternally expressed like its orthologues in human, sheep, and mouse. Conclusion This is the first report on the imprinting status of SLC38A4, NAP1L5, and on the expression patterns of the two transcripts of NNAT in cattle. It is of interest that the imprinting of NAP1L5, NNAT, and H19 appears to be conserved between mouse and cow, although the tissue distribution of expression differs. In contrast, the imprinting of SLC38A4 appears to be species-specific.
Collapse
Affiliation(s)
- Ismail Zaitoun
- Department of Dairy Science, University of Wisconsin-Madison, 1675 Observatory Dr., Madison, WI 53706, USA
| | - Hasan Khatib
- Department of Dairy Science, University of Wisconsin-Madison, 1675 Observatory Dr., Madison, WI 53706, USA
| |
Collapse
|
118
|
Gebhard C, Schwarzfischer L, Pham TH, Schilling E, Klug M, Andreesen R, Rehli M. Genome-Wide Profiling of CpG Methylation Identifies Novel Targets of Aberrant Hypermethylation in Myeloid Leukemia. Cancer Res 2006; 66:6118-28. [PMID: 16778185 DOI: 10.1158/0008-5472.can-06-0376] [Citation(s) in RCA: 157] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The methylation of CpG islands is associated with transcriptional repression and, in cancer, leads to the abnormal silencing of tumor suppressor genes. Because aberrant hypermethylation may be used as a marker for disease, a sensitive method for the global detection of DNA methylation events is of particular importance. We describe a novel and robust technique, called methyl-CpG immunoprecipitation, which allows the unbiased genome-wide profiling of CpG methylation in limited DNA samples. The approach is based on a recombinant, antibody-like protein that efficiently binds native CpG-methylated DNA. In combination with CpG island microarrays, the technique was used to identify >100 genes with aberrantly methylated CpG islands in three myeloid leukemia cell lines. Interestingly, within all hypermethylation targets, genes involved in transcriptional regulation were significantly overrepresented. More than half of the identified genes were absent in microarray expression studies in either leukemia or normal monocytes, indicating that hypermethylation in cancer may be largely independent of the transcriptional status of the affected gene. Most individually tested genes were also hypermethylated in primary blast cells from acute myeloid leukemia patients, suggesting that our approach can identify novel potential disease markers. The technique may prove useful for genome-wide comparative methylation analysis not only in malignancies.
Collapse
Affiliation(s)
- Claudia Gebhard
- Department of Hematology and Oncology, University Hospital, Regensburg, Germany
| | | | | | | | | | | | | |
Collapse
|
119
|
Isles AR, Humby T. Modes of imprinted gene action in learning disability. JOURNAL OF INTELLECTUAL DISABILITY RESEARCH : JIDR 2006; 50:318-25. [PMID: 16629925 DOI: 10.1111/j.1365-2788.2006.00843.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
BACKGROUND It is now widely acknowledged that there may be a genetic contribution to learning disability and neuropsychiatric disorders, stemming from evidence provided by family, twin and adoption studies, and from explicit syndromic conditions. Recently it has been recognized that in some cases the presentation of genetic syndromes (or discrete aspects of disorders) is dependent on the sex of the transmitting parent. Such 'parent-of-origin effects' can be explained by a number of genetic mechanisms, a predominant one of which is genomic imprinting. Genomic imprinting refers to the parent of origin-specific epigenetic marking of an allele of a gene, such that for some genes it is mainly the maternally inherited allele only that is expressed, whereas for others expression occurs mainly from the paternal copy. METHODS Here we discuss the contribution of imprinted genes to mental dysfunction and learning disability, using clinical examples of association studies and explicit imprinting disorders (with particular emphasis to Angelman and Prader-Willi syndromes), and evidence from animal work. RESULTS Clinical and animal studies strongly suggest that imprinted genes contribute to brain functioning, and when the genes or epigenetic processes are disrupted, this can give rise to neuropsychiatric problems. Another system to which imprinted genes provide a large contribute is the placenta and foetal development. Epidemiological studies suggest that this is also a key area in which dysregulation can give rise to learning difficulties. CONCLUSIONS Disruption of imprinted genes, or the epigenetic processes controlling them, can contribute to learning disability. These effects can be divided into two types: direct effects, such as those seen in explicit imprinting disorders such as Angelman and Prader-Willi syndromes, and indirect effects as manifest via changes in foetal programming.
Collapse
|
120
|
Maeda N, Hayashizaki Y. Genome-wide survey of imprinted genes. Cytogenet Genome Res 2006; 113:144-52. [PMID: 16575174 DOI: 10.1159/000090826] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2005] [Accepted: 09/15/2005] [Indexed: 01/06/2023] Open
Abstract
The developmental failure of mammalian parthenogenote has been a mystery for a long time and posed a question as to why bi-parental reproduction is necessary for development to term. In the 1980s, it was proven that this failure was not due to the genetic information itself, but to epigenetic modification of genomic DNA. In the following decade, several studies successfully identified imprinted genes which were differentially expressed in a parent-of-origin-specific manner, and it was shown that the differential expression depended on the pattern of DNA methylation. These facts prompted development of genome-wide systematic screening methods based on DNA methylation and differential gene expression to identify imprinted genes. Recently computational approaches and microarray technology have been introduced to identify imprinted genes/loci, contributing to the expansion of our knowledge. However, it has been shown that the gene silencing derived from genomic imprinting is accomplished by several mechanisms in addition to direct DNA methylation, indicating that novel approaches are further required for comprehensive understanding of genomic imprinting. To unveil the mechanism of developmental failure in mammalian parthenogenote, systematic screenings for imprinted genes/loci have been developed. In this review, we describe genomic imprinting focusing on the history of genome-wide screening.
Collapse
Affiliation(s)
- N Maeda
- Genome Science Laboratory, Discovery and Research Institute, RIKEN, Saitama, Japan
| | | |
Collapse
|
121
|
Ruf N, Dünzinger U, Brinckmann A, Haaf T, Nürnberg P, Zechner U. Expression profiling of uniparental mouse embryos is inefficient in identifying novel imprinted genes. Genomics 2006; 87:509-19. [PMID: 16455231 DOI: 10.1016/j.ygeno.2005.12.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2005] [Revised: 12/15/2005] [Accepted: 12/15/2005] [Indexed: 12/23/2022]
Abstract
Imprinted genes are expressed from only one allele in a parent-of-origin-specific manner. We here describe a systematic approach to identify novel imprinted genes using quantification of allele-specific expression by Pyrosequencing, a highly accurate method to detect allele-specific expression differences. Sixty-eight candidate imprinted transcripts mapping to known imprinted chromosomal regions were selected from a recent expression profiling study of uniparental mouse embryos and analyzed. Three novel imprinted transcripts encoding putative non-protein-coding RNAs were identified on the basis of parent-of-origin-specific monoallelic expression in E11.5 (C57BL/6 x Cast/Ei)F1 and informative (C57BL/6 x Cast/Ei) x C57BL/6 backcross embryos. In addition, four transcripts with preferential expression of a strain-specific allele were found. Intriguingly, a vast majority of the analyzed transcripts showed no imprinting-associated expression in F1 embryos. These data strengthen the view that a large fraction of nonimprinted genes is differentially expressed between parthenogenetic and androgenetic embryos and question the efficiency of expression profiling of uniparental embryos to identify novel imprinted genes.
Collapse
MESH Headings
- Alleles
- Animals
- Chromosome Mapping
- Chromosomes
- Crosses, Genetic
- DNA/genetics
- DNA/isolation & purification
- DNA, Complementary/genetics
- Databases, Genetic
- Embryo, Mammalian
- Female
- Gene Expression
- Gene Expression Profiling
- Genetic Variation
- Genomic Imprinting
- Mice
- Mice, Inbred C57BL
- Mice, Inbred Strains
- Models, Genetic
- Parthenogenesis
- Polymorphism, Single Nucleotide
- Prader-Willi Syndrome/genetics
- Pregnancy
- RNA/isolation & purification
- RNA, Messenger/genetics
- Software
- Transcription, Genetic
Collapse
Affiliation(s)
- Nico Ruf
- Max-Delbrueck-Center for Molecular Medicine, Berlin-Buch, Germany
| | | | | | | | | | | |
Collapse
|
122
|
Constância M, Angiolini E, Sandovici I, Smith P, Smith R, Kelsey G, Dean W, Ferguson-Smith A, Sibley CP, Reik W, Fowden A. Adaptation of nutrient supply to fetal demand in the mouse involves interaction between the Igf2 gene and placental transporter systems. Proc Natl Acad Sci U S A 2005; 102:19219-24. [PMID: 16365304 PMCID: PMC1316882 DOI: 10.1073/pnas.0504468103] [Citation(s) in RCA: 237] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The mammalian fetus is unique in its dependence during gestation on the supply of maternal nutrients through the placenta. Maternal supply and fetal demand for nutrients need to be fine tuned for healthy growth and development of the fetus along its genetic trajectory. An altered balance between supply and demand can lead to deviations from this trajectory with long-term consequences for health. We have previously shown that in a knockout lacking the imprinted placental-specific Igf2 transcript (P0), growth of the placenta is compromised from early gestation but fetal growth is normal until late gestation, suggesting functional adaptation of the placenta to meet the fetal demands. Here, we show that placental transport of glucose and amino acids are increased in the Igf2 P0(+/-) null and that this up-regulation of transport occurs, at least in part, through increased expression of the transporter genes Slc2a3 and Slc38a4, the imprinted member of the System A amino acid transporter gene family. Decreasing fetal demand genetically by removal of fetal Igf2 abolished up-regulation of both transport systems and reduced placental System A amino acid transport activity and expression of Slc38a2 in late gestation. Our results provide direct evidence that the placenta can respond to fetal demand signals through regulation of expression of specific placental transport systems. Thus, crosstalk between an imprinted growth demand gene (Igf2) and placental supply transporter genes (Slc38a4, Slc38a2, and Slc2a3) may be a component of the genetic control of nutrient supply and demand during mammalian development.
Collapse
Affiliation(s)
- Miguel Constância
- Laboratory of Developmental Genetics and Imprinting, The Babraham Institute, Babraham Research Campus, Cambridge CB2 4AT, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Sibley CP, Turner MA, Cetin I, Ayuk P, Boyd CAR, D'Souza SW, Glazier JD, Greenwood SL, Jansson T, Powell T. Placental phenotypes of intrauterine growth. Pediatr Res 2005; 58:827-32. [PMID: 16183820 DOI: 10.1203/01.pdr.0000181381.82856.23] [Citation(s) in RCA: 172] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The placenta is essential to nutrition before birth. Recent work has shown that a range of clearly defined alterations can be found in the placentas of infants with intrauterine growth restriction (IUGR). In the mouse, a placental specific knockout of a single imprinted gene, encoding IGF-2, results in one pattern of alterations in placenta structure and function which leads to IUGR. We speculate that the alterations in the human placenta can also be grouped into patterns, or phenotypes, that are associated with specific patterns of fetal growth. Identifying the placental phenotypes of different fetal growth patterns will improve the ability of clinicians to recognize high-risk patients, of laboratory scientists to disentangle the complexities of IUGR, and of public health teams to target interventions aimed at ameliorating the long-term adverse effects of inadequate intrauterine growth.
Collapse
Affiliation(s)
- Colin P Sibley
- Division of Human Development, Acadamic Unit of Child Health, The Medical School, University of Manchester, St. Mary's Hospital, Manchester M13 OJH, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
124
|
Youngson NA, Kocialkowski S, Peel N, Ferguson-Smith AC. A Small Family of Sushi-Class Retrotransposon-Derived Genes in Mammals and Their Relation to Genomic Imprinting. J Mol Evol 2005; 61:481-90. [PMID: 16155747 DOI: 10.1007/s00239-004-0332-0] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2004] [Accepted: 03/29/2005] [Indexed: 02/08/2023]
Abstract
Ty3/gypsy retrotransposons are rare in mammalian genomes despite their abundance in invertebrate and other vertebrate classes. Here we identify a family of nine conserved mammalian genes with homology to Ty3/gypsy retrotransposons but which have lost their ability to autonomously retrotranspose. Of these, five map to the X chromosome while the remaining four are autosomal. Comparative phylogenetic analyses show them to have strongest homology to the sushi-ichi element from Fugu rubripes. Two of the autosomal gene members, Peg10 and Rtl1, are known to be imprinted, being expressed from the paternally inherited chromosome homologue. This suggests, consistent with the host-parasite response theory of the evolution of the imprinting mechanism, that parental-origin specific epigenetic control may be mediated by genomic "parasitic" elements such as these. Alternatively, these elements may preferentially integrate into regions that are differentially modified on the two homologous chromosomes such as imprinted domains and the X chromosome and acquire monoallelic expression. We assess the imprinting status of the remaining autosomal members of this family and show them to be biallelically expressed in embryo and placenta. Furthermore, the methylation status of Rtl1 was assayed throughout development and was found to resemble that of actively, silenced repetitive elements rather than imprinted sequences. This indicates that the ability to undergo genomic imprinting is not an inherent property of all members of this family of retroelements. Nonetheless, the conservation but functional divergence between the different members suggests that they have undergone positive selection and acquired distinct endogenous functions within their mammalian hosts.
Collapse
Affiliation(s)
- Neil A Youngson
- Department of Anatomy, University of Cambridge, Downing St, Cambridge, CB23DY, UK
| | | | | | | |
Collapse
|
125
|
Desforges M, Lacey HA, Glazier JD, Greenwood SL, Mynett KJ, Speake PF, Sibley CP. SNAT4 isoform of system A amino acid transporter is expressed in human placenta. Am J Physiol Cell Physiol 2005; 290:C305-12. [PMID: 16148032 DOI: 10.1152/ajpcell.00258.2005] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The system A amino acid transporter is encoded by three members of the Slc38 gene family, giving rise to three subtypes: Na+-coupled neutral amino acid transporter (SNAT)1, SNAT2, and SNAT4. SNAT2 is expressed ubiquitously in mammalian tissues; SNAT1 is predominantly expressed in heart, brain, and placenta; and SNAT4 is reported to be expressed solely by the liver. In the placenta, system A has an essential role in the supply of neutral amino acids needed for fetal growth. In the present study, we examined expression and localization of SNAT1, SNAT2, and SNAT4 in human placenta during gestation. Real-time quantitative PCR was used to examine steady-state levels of system A subtype mRNA in early (6-10 wk) and late (10-13 wk) first-trimester and full-term (38-40 wk) placentas. We detected mRNA for all three isoforms from early gestation onward. There were no differences in SNAT1 and SNAT2 mRNA expression with gestation. However, SNAT4 mRNA expression was significantly higher early in the first trimester compared with the full-term placenta (P < 0.01). We next investigated SNAT4 protein expression in human placenta. In contrast to the observation for gene expression, Western blot analysis revealed that SNAT4 protein expression was significantly higher at term compared with the first trimester (P < 0.05). Immunohistochemistry and Western blot analysis showed that SNAT4 is localized to the microvillous and basal plasma membranes of the syncytiotrophoblast, suggesting a role for this isoform of system A in amino acid transport across the placenta. This study therefore provides the first evidence of SNAT4 mRNA and protein expression in the human placenta, both at the first trimester and at full term.
Collapse
Affiliation(s)
- M Desforges
- Division of Human Development, St. Mary's Hospital, The Medical School, University of Manchester, Manchester, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
126
|
Abstract
The success of the human genome sequencing project has created wide-spread interest in exploring the human epigenome in order to elucidate how the genome executes the information it holds. Although all (nucleated) human cells effectively contain the same genome, they contain very different epigenomes depending upon cell type, developmental stage, sex, age and various other parameters. This complexity makes it intrinsically difficult to precisely define 'an' epigenome, let alone 'the' epigenome. What is clear, however, is that in order to unravel any epigenome, existing and novel high-throughput approaches on the DNA, RNA and protein levels need to be harnessed and integrated. Here, we review the current thinking and progress on how to get from the genome to the epigenome(s) and discuss some potential applications.
Collapse
Affiliation(s)
- Adele Murrell
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK.
| | | | | |
Collapse
|
127
|
Abstract
Imprinted genes are expressed monoallelically depending on their parental origin. High expression of the majority of imprinted genes tested to date has been demonstrated in extraembryonic tissues; placenta and yolk sac. Several mouse models where specific imprinted genes have been disrupted demonstrate that fetal and placental growth may be regulated by imprinted genes, in which paternally expressed genes enhance, and maternally expressed genes restrain, growth. We review the current information on, and suggest possible functional roles for, imprinted genes in placental development.
Collapse
Affiliation(s)
- P M Coan
- Department of Anatomy, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK.
| | | | | |
Collapse
|
128
|
Russanova VR, Hirai TH, Howard BH. Semirandom sampling to detect differentiation-related and age-related epigenome remodeling. J Gerontol A Biol Sci Med Sci 2005; 59:1221-33. [PMID: 15699521 DOI: 10.1093/gerona/59.12.1221] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
With completion of the human genome project, patterns of higher order chromatin structure can be easily related to other features of genome organization. A well-studied aspect of chromatin, histone H4 acetylation, is examined here on the basis of its role in setting competence for gene activation. Three applications of a new hybrid genome sampling-chromatin immunoprecipitation strategy are described. The first explores aspects of epigenome architecture in human fibroblasts. A second focuses on chromatin from HL-60 promyelocytic leukemia cells before and after differentiation into macrophage-like cells. A third application explores age-related epigenome change. In the latter, acetylation patterns are compared in human skin fibroblast chromatin from donors of various ages. Two sites are reported at which observed histone H4 acetylation differences suggest decreasing acetylation over time. The sites, located in chromosome 4p16.1 and 4q35.2 regions, appear to remodel during late fetal-early child development and from preadolescence through adult life, respectively.
Collapse
Affiliation(s)
- Valya R Russanova
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
129
|
Abstract
Studies in mouse and human have established that the exon encoding the neuroendocrine secretory protein Nesp55 is transcribed exclusively from the maternal allele. To study the imprinting status of Nesp55 in cattle, we examined transcripts of this exon in a wide range of tissues from four fetuses and the dam of one of the fetuses (not all tissues were examined in all subjects). A polymorphic site was used to determine transcript status as monoallelic or biallelic. The expression analysis was by direct sequencing of RT-PCR products amplified from the tissues. Nesp55 transcripts were detected in many tissues, in contrast to previous studies in which Nesp55 was detected by immunoassays in endocrine and nervous tissues only. In all five individuals, Nesp55 showed monoallelic expression. In one of the fetuses, parental origin of the expressed allele could not be determined; in the others monoallelic expression was clearly maternal. Thus, bovine Nesp55 is maternally expressed like its counterparts in mouse and human.
Collapse
Affiliation(s)
- Hasan Khatib
- Department of Dairy Science, University of Wisconsin, 1675 Observatory Drive, Madison 53706, USA.
| |
Collapse
|
130
|
Brandt J, Veith AM, Volff JN. A family of neofunctionalized Ty3/gypsy retrotransposon genes in mammalian genomes. Cytogenet Genome Res 2005; 110:307-17. [PMID: 16093683 DOI: 10.1159/000084963] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2003] [Accepted: 02/25/2004] [Indexed: 12/30/2022] Open
Abstract
A family of at least eleven genes called Mar related to long terminal repeat retrotransposons from the Ty3/gypsy group, including two genes previously identified as such, is present in human and mouse genomes. Single orthologous copies were identified for most Mar genes in different mammals. All of them have lost essential structural features necessary for autonomous retrotransposition before divergence between mouse and human. Three Mar genes also have introns at identical positions in human and mouse. Hence, Mar genes do not correspond to functional retrotransposons. Mar genes evolved under purifying selection, strongly suggesting that they are not pseudogenic relics but rather neofunctionalized retrotransposon genes. All putative Mar proteins display sequence similarity to the capsid-like domain of the Gag protein of Tf1/Sushi retrotransposons. In addition, three Mar proteins have conserved the Gag CCHC zinc finger motif, suggesting a role in nucleic acid binding. Some Mar genes have also retained from their retrotransposon origin a -1 ribosomal frameshifting between the gag-related open reading frame and a region encoding a putative aspartyl protease domain. EST analysis revealed that the majority of Mar genes are expressed in brain as well as in other tissues and organs. Some Mar proteins might function as transcription factors or be involved in the control of cell proliferation and apoptosis. Strikingly, as many as eight Mar genes are located on the X chromosome in human, mouse and other mammals, and at least two of the autosomal genes are subject to imprinting. We suggest that retrotransposons might be a source for epigenetically regulated genes. Epigenetic regulation of these neogenes might be derived from the cellular defense mechanisms having controlled their retrotransposon ancestor.
Collapse
Affiliation(s)
- J Brandt
- Biofuture Research Group, Physiologische Chemie I, Biozentrum, University of Würzburg, Würzburg, Germany
| | | | | |
Collapse
|
131
|
Davies W, Smith RJ, Kelsey G, Wilkinson LS. Expression patterns of the novel imprinted genes Nap1l5 and Peg13 and their non-imprinted host genes in the adult mouse brain. Gene Expr Patterns 2004; 4:741-7. [PMID: 15465498 DOI: 10.1016/j.modgep.2004.03.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2004] [Revised: 03/19/2004] [Accepted: 03/20/2004] [Indexed: 01/10/2023]
Abstract
Recent work has implicated imprinted gene functioning in neurodevelopment and behaviour and defining the expression patterns of these genes in brain tissue has become a key prerequisite to establishing function. In this work we report on the expression patterns of two novel imprinted loci, Nap1l5 and Peg13, in adult mouse brain using in situ hybridisation methods. Nap1l5 and Peg13 are located, respectively, within the introns of the non-imprinted genes Herc3 and the Tularik1 (T1)/KIAA1882 homologue in two separate microimprinted domains on mouse chromosomes 6 and 15. These 'host' genes are highly expressed in brain and consequently we were interested in assessing their expression patterns in parallel to the imprinted genes. The brain expression of all four genes appeared to be mainly neuronal. The detailed expression profiles of Nap1l5 and Peg13 were generally similar with widespread expression that was relatively high in the septal and hypothalamic regions, the hippocampus and the cerebral cortex. In contrast, there was some degree of dissociation between the imprinted genes and their non-imprinted hosts, in that, whilst there was again widespread expression of Herc3 and the T1/KIAA1882 homologue, these genes were also particularly highly expressed in Purkinje neurons and piriform cortex. We also examined expression of the novel imprinted genes in the adrenal glands. Nap1l5 expression was localised mainly to the adrenal medulla, whilst Peg13 expression was observed more generally throughout the adrenal medulla and the outer cortical layers.
Collapse
Affiliation(s)
- William Davies
- Developmental Genetics and Neurobiology Programmes, The Babraham Institute, Babraham, Cambridge CB2 4AT, UK
| | | | | | | |
Collapse
|
132
|
Beechey CV. A reassessment of imprinting regions and phenotypes on mouse chromosome 6: Nap1l5 locates within the currently defined sub-proximal imprinting region. Cytogenet Genome Res 2004; 107:108-14. [PMID: 15305064 DOI: 10.1159/000079579] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2004] [Accepted: 04/05/2004] [Indexed: 11/19/2022] Open
Abstract
Previous studies (Beechey, 2000) have shown that mouse proximal chromosome (Chr) 6 has two imprinting regions. An early embryonic lethality is associated with two maternal copies of the more proximal imprinting region, while mice with two maternal copies of the sub-proximal imprinting region are growth retarded at birth, the weight reduction remaining similar to adulthood. No detectable postnatal imprinting phenotype was seen in these earlier studies with two paternal copies of either region. The sub-proximal imprinting region locates distal to the T77H reciprocal translocation breakpoint in G-band 6A3.2 and results reported here show that it does not extend beyond the breakpoint of the more distal T6Ad translocation in 6C2. It has been confirmed that the postnatal growth retardation observed with two maternal copies of the sub-proximal region is established in utero, although placental size was normal. A new finding is that 16.5-18.5-dpc embryos, with two paternal copies of the sub-proximal imprinting region, were larger than their normal sibs, although placental size was normal. As no postnatal growth differences have been observed in these mice, the fetal overgrowth must normalize by birth. The imprinted genes Peg1/Mest, Copg2, Copg2as and Mit1/Lb9 map to the sub-proximal imprinting region and are thus candidates for the observed imprinting phenotypes. Another candidate is the recently reported imprinted gene Nap1l5. Expression studies of Nap1l5 in mice with two maternal or two paternal copies of different regions of Chr 6 have demonstrated that the gene locates within the sub-proximal imprinting region. FISH has mapped Nap1l5 to G-band 6C1, within the sub-proximal imprinting region but several G-bands distal to the Peg1/Mest cluster. This location, and the 30-Mb separation of these loci on the sequence map, makes it probable that Nap1l5 defines a new imprinting domain within the currently defined sub-proximal imprinting region.
Collapse
Affiliation(s)
- C V Beechey
- Mammalian Genetics Unit, Medical Research Council, Harwell, Didcot, Oxfordshire, UK.
| |
Collapse
|
133
|
Smith RJ, Arnaud P, Kelsey G. Identification and properties of imprinted genes and their control elements. Cytogenet Genome Res 2004; 105:335-45. [PMID: 15237221 DOI: 10.1159/000078206] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2003] [Accepted: 12/10/2003] [Indexed: 11/19/2022] Open
Abstract
Imprinted genes have the unusual characteristic that the copy from one parent is destined to remain inactive. Though few in number they nonetheless constitute a functionally important part of the mammalian genome. With their memory of parental origin, imprinted genes represent an important model for the epigenetic regulation of gene function and will provide invaluable paradigms to test whether we can predict epigenetic state from DNA sequence. Since their first discovery, systematic screens and some good fortune have led to identification of over seventy imprinted genes in the mouse and human: recent microarray analysis may reveal many more. With a significant number of imprinted genes now identified and completion of key mammalian genome sequences, we are able systematically to examine the organization of imprinted loci, properties of their control elements and begin to recognize common themes in imprinted gene regulation.
Collapse
Affiliation(s)
- R J Smith
- Developmental Genetics Programme, The Babraham Institute, Cambridge, UK
| | | | | |
Collapse
|
134
|
Wang Y, Joh K, Masuko S, Yatsuki H, Soejima H, Nabetani A, Beechey CV, Okinami S, Mukai T. The mouse Murr1 gene is imprinted in the adult brain, presumably due to transcriptional interference by the antisense-oriented U2af1-rs1 gene. Mol Cell Biol 2004; 24:270-9. [PMID: 14673161 PMCID: PMC303337 DOI: 10.1128/mcb.24.1.270-279.2004] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The mouse Murr1 gene contains an imprinted gene, U2af1-rs1, in its first intron. U2af1-rs1 shows paternal allele-specific expression and is transcribed in the direction opposite to that of the Murr1 gene. In contrast to a previous report of biallelic expression of Murr1 in neonatal mice, we have found that the maternal allele is expressed predominantly in the adult brain and also preferentially in other adult tissues. This maternal-predominant expression is not observed in embryonic and neonatal brains. In situ hybridization experiments that used the adult brain indicated that Murr1 gene was maternally expressed in neuronal cells in all regions of the brain. We analyzed the developmental change in the expression levels of both Murr1 and U2af1-rs1 in the brain and liver, and we propose that the maternal-predominant expression of Murr1 results from transcriptional interference of the gene by U2af1-rs1 through the Murr1 promoter region.
Collapse
Affiliation(s)
- Youdong Wang
- Department of Biomolecular Sciences, Saga Medical School, Saga 849-8501, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
135
|
Walter J, Paulsen M. The potential role of gene duplications in the evolution of imprinting mechanisms. Hum Mol Genet 2003; 12 Spec No 2:R215-20. [PMID: 12944422 DOI: 10.1093/hmg/ddg296] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Using the completed genomic sequences of mouse and human we performed a comparative analyses of imprinted genes and gene clusters. For many imprinted genes we could detect imprinted as well as non-imprinted paralogues. The inter- and intrachromosomal similarities between paralogues and their linkage to imprinting clusters suggests that imprinted genes were dispersed throughout the genome by gene duplications as well as translocation and transposition events. Our findings indicate that imprinting clusters may have been linked together on one (or a few) ancestral pre-imprinted chromosome(s), arguing for a common mechanistic origin of imprinting control. Imprinting may originally have evolved on a simple basis of dosage compensation required for some duplicated genes (chromosomes) followed by selection of sex-biased expression control.
Collapse
Affiliation(s)
- Jorn Walter
- Universität des Saarlandes, Saarbrücken, Germany
| | | |
Collapse
|